Next Article in Journal
Alcohol as a Modifiable Risk Factor for Alzheimer’s Disease—Evidence from Experimental Studies
Previous Article in Journal
Enzymatic Hydrolysis Optimization for Preparation of Sea Cucumber (Holothuria scabra) Hydrolysate with an Antiproliferative Effect on the HepG2 Liver Cancer Cell Line and Antioxidant Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Design, Synthesis and Biological Activities of (Thio)Urea Benzothiazole Derivatives

by
Jessica E. Mendieta-Wejebe
1,
Martha C. Rosales-Hernández
1,
Itzia I. Padilla-Martínez
2,
Efrén V. García-Báez
2 and
Alejandro Cruz
2,*
1
Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de Mexico 11340, Mexico
2
Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Barrio la Laguna Ticomán, Ciudad de Mexico 07340, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9488; https://doi.org/10.3390/ijms24119488
Submission received: 9 March 2023 / Revised: 17 May 2023 / Accepted: 24 May 2023 / Published: 30 May 2023
(This article belongs to the Section Molecular Pharmacology)

Abstract

:
(Thio)ureas ((T)Us) and benzothiazoles (BTs) each have demonstrated to have a great variety of biological activities. When these groups come together, the 2-(thio)ureabenzothizoles [(T)UBTs] are formed, improving the physicochemical as well as the biological properties, making these compounds very interesting in medicinal chemistry. Frentizole, bentaluron and methabenzthiazuron are examples of UBTs used for treatment of rheumatoid arthritis and as wood preservatives and herbicides in winter corn crops, respectively. With this antecedent, we recently reported a bibliographic review about the synthesis of this class of compounds, from the reaction of substituted 2-aminobenzothiazoles (ABTs) with iso(thio)cyanates, (thio)phosgenes, (thio)carbamoyl chlorides, 1,1’-(thio)carbonyldiimidazoles, and carbon disulfide. Herein, we prepared a bibliographic review about those features of design, chemical synthesis, and biological activities relating to (T)UBTs as potential therapeutic agents. This review is about synthetic methodologies generated from 1968 to the present day, highlighting the focus to transform (T)UBTs to compounds containing a range substituents, as illustrated with 37 schemes and 11 figures and concluded with 148 references. In this topic, the scientists dedicated to medicinal chemistry and pharmaceutical industry will find useful information for the design and synthesis of this interesting group of compounds with the aim of repurposing these compounds.

1. Introduction

(Thio)urea ((T)U) is a valuable functional group present in molecular structures used in several areas of chemistry and drug design. Recently, (T)U compounds have gained the attention of several researchers due to their chemical and biological properties. Due to the NH hydrogen atoms and their ability to form hydrogen bonds with the target substrates, TUs have been used in the design of molecules of biological interest. They show a broad array in their biological properties, such as anti-HIV [1], analgesic [2], antibacterial [3,4,5], antimicrobial [6,7,8,9,10,11,12,13], anticancer [14,15,16,17,18], antifungal [19,20], diuretic [21], antiviral [22,23], anticonvulsant [24], anti-thyroidal [25], herbicidal and insecticidal [26], anti-inflammatory [27], anti-acetylcholinesterase [28,29], anti-tuberculosis [30,31], antimalarial [32,33], hypoglycemic [34], CCR4 antagonists [35], and DNA-topoisomerase inhibitors [36].
Among drugs used in current medicine, some of them have a benzothiazole (BT) nucleus. BT core was found to be in natural compounds with biological activities. Since the pharmacological profiling of riluzole (6-trifluoromethoxy-benzothiazole-2-amine) became known as a clinically anticonvulsant drug in 1950 [37,38], medicinal chemists have been interested in BT derivatives. The development in biological evaluation of this heterocyclic compounds has initiated the design of novel molecules based on their mechanism of action. Nowadays, BT itself is a very important phamacophore in medicinal chemistry due to the broad pharmacological properties of its derivatives. For instance, from the period 2000–2010, only five scientific reviews were found in the literature, briefly describing important findings on synthetic methodologies and medicinal activities associated with BT compounds [39,40,41,42]. However, in the past decade, the use of BT core for drug development, has been increased rapidly. Thirty reviews were found that focused on research highlighting anticancer, antimicrobial, anticonvulsant, anti-inflammatory, antifungal, anti-oxidant, anti-tubercular, anti-malarial, anti-leishmanial, anti-Alzheimer, anti-diabetic, and other miscellaneous activities [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72]. Some BTs are used as pharmaceuticals for the treatment of diseases such as cancer, neurodegenerative disorders, Huntington’s disease, and local brain ischemia. In the same period, an article review for the period from 2015 to 2020, relating to patents of pharmacological activities of BTs, was reported [73]. Even from the last past year until now, thirteen reviews about this topic have been reported in the literature [74,75,76,77,78,79,80,81,82,83,84,85].
Nowadays, scientists are fighting to find drugs against dangerous viruses for human health. For instance, human immunodeficiency, poliovirus, and coronavirus has destroyed many lives. Scientists have found many antiviral drugs per type of virus. From them, BT derivatives have been proved to act as good antiviral agents. Researchers are continually working on the use of BT moiety to obtain best BT derivatives to eventually be used as antiviral agents. Recently, two article reviews related to the synthesis, structure–activity relationship, and several methods for evaluating the antiviral activity against specific viruses of BT derivatives were reported [86,87]. For instance, the frentizole drug (Figure 1), a nontoxic UBT used to treat the rheumatoid arthritis and systemic lupus erythematosus were studied for the immunosuppressive and super-immunosuppressive dose levels on the resistance of the mice to viral infections. It was found that frentizol reduced the herpes simplex and influence virus. Additionally, it is known that various (T)UBTs inhibit DNA topoisomerase or HIV reverse transcriptase (e.g., I, Figure 1) [88,89,90]. Additionally, bentaluron and methabenzthiazuron are fungicides used as a wood preservative and herbicide in winter corn crops, respectively [91,92]. In addition, among the drugs discovered in the recent years, several (T)UBTs are potential drugs for the Alzheimer′s disease treatment [93,94,95,96,97,98].
The broad spectrum of biological activity of (T)U as well as BT derivatives encouraged us to review the methods of synthesis of (T)UBTs and their biological activity studies. In 2022, we published a literature review relating to the research progress on the synthesis and biological importance of (T)UBTs. In that work, we summarized the methodologies to synthetize (T)UBTs from the condensation of substituted 2ABTs with iso(thio)cyanates, (thio)phosgenes, 1,10-(thio)carbonyldiimidazoles (thio)carbodiimides, (T)CDIs, (thio)carbamoyl chlorides, and carbon disulfide as starting materials, covering the last 40 years [99]. In view of the fact that the synthesis and biological activity of new 2-(T)UBT derivatives have been considerably increased, and in continuation with our research in this work, we prepared a bibliographic review about these kinds of compounds; this work relates to the synthetic procedures to access (T)UBT derivatives, starting from (T)UBT as starting materials and their analysis regarding to their biological activities, from 1968 to the present day.
The work was divided into reaction methodologies starting with (un)substituted-2ABT to afford the respective (T)UBT, which were transformed into (1) (T)UBT derivatives, (2) BT-semi(thio)carbazide derivatives, (3) N-acyl-(T)UBT derivatives, and (4) GBT derivatives.

2. (T)UBT Derivatives

N-(4,6-disubstituted-BT-2-yl)-N′-(β-bromopropionyl)ureas 1ag or the N-(6-disubstituted-BT-2-yl)- N′-(acryloyl)ureas 2a,b were prepared from the reaction between 4,6-disubstituted-2ABT and β-bromopropionyl isocyanate or acryloylisothiocyanate, respectively, Scheme 1 [100]. The ureas 1d,e and 2a,b were cyclicized to 1-(6-disubstituted-benzothiazol-2-yl)dihydrouracils 3a,b, respectively. All compounds were analyzed in vitro against antibacterial, antifungal, and antiprotozoal effects. The minimal inhibition concentration of 6-thiocyanate-N-β-bromopropionyl-UBT 1g was 50 μg/mL for all tested organisms. However, the minimal inhibition concentrations of dihydrouracil-BT 3a and 3b were higher for all tested organisms.
The N-(4-methyl-BT-2-yl)-N′-(BI-N-sulfonyl)urea 4 was prepared from the reaction of BI-N-sulfonyl chloride and 4-methyl-UBT as starting materials, Scheme 2 [101]. The antitumor activity of this compound showed GI50, TGI, and LC50 values of 25.1, 77.5, and 93.3 μM, respectively. Compound 4 had a broad-spectrum antitumor activity and selective activity to single cell lines. Distinctive activities were showed compared to that of sulofenur against EKVX non-small lung cancer, RPMI-8226 leukemia, OVCAR-4 ovarian cancer, PC-3 prostate cancer, CAKI-1 renal cancer, MDA-MB-435, and T-47D breast cancer with GI50 values of 1.7, 21.5, 25.9, 28.7, 15.9, 27.9, and 15.1 μM, respectively.
Nine N-(4/6-substituted-BT-2-yl)-N′-(phenyl)thioureas 5, obtained from 4/6-substituted 2ABTs and phenylisothiocyanate in refluxing ethanol, were reacted with malonic acid in presence of acetyl chloride to synthesize 1-(4/6-substituted-BT-2-yl)-3-phenyl-2-thiobarbituric acids 6ai, Scheme 3 [102]. Compounds 5 and 6 were screened for their entomological and antibacterial activities. They were tested for antibacterial activity at 200 μg/mL and 100 μ/mL in DMF against Gram-(+) and Gram-(−) bacteria, using streptomycin and ceftazidime as reference drugs. Compounds 6a, 6f, and 6g showed broad-spectrum activities, but they were less active than the reference drugs. The same compounds had potent antiulcer, anti-inflammatory, antitumor, antifeedant, and acaricidal activities against Spodoptera litura and Tetranychus urticae, respectively. These compounds was proposed to be better used in drug development for bacterial infections, antifeedant, and acaricidal activities in the future as well.
N-(6-cyano-BT-2-yl)-N′-(p-MeObenzyl)urea 7, obtained from 6-cyano-2ABT and the corresponding benzylisocyanate, was utilized in the generation of N-(6-tetrazolyl-BT-2-yl)-N′-(p-methoxy-benzyl)urea 8 in 91% yield using microwave irradiation, Scheme 4 [103]. The arylureas 7 and 8 were evaluated for their inhibition of GSK-3 activity. Compound 8 was found to reduce in vitro the GSK-3β activity beneath 57% at a concentration of 1.0 μM. The structure–activity relationship of the library provided the rationale for the synthesis of UBT 8 (IC50 = 140 nM), which displayed more than a two-fold increase in activity compared with the standard reference AR-A014418 (IC50 = 330 nM).
A series of twelve N-(4/6-substituted-BT-2-yl)-N′-(p-substituted-aryl)ureas 9al was synthetized for its chemoselective oxidative cyclization with 1,3-di-n-butylimidazolium tribromide [bbim-Br3] to yield the N-bis-BTs 10al, Scheme 5 [91]. The cytotoxic activity against a mouse melanoma cell line (B16-F10) and two human monocytic cell lines (U937, THP-1) of compounds 9 and 10 were evaluated. The compounds 9b, 9e, 9f, 9k, 9c, and 10h were the most active based on their IC50 values. Among them, compound 9e (16.23 μM) showed the more antiproliferative activity on U937 cells compared with the reference, etoposide (17.94 μM). Nevertheless, these compounds showed less cytotoxicity towards THP-1 cells.
N-(6-Methoxy-BT-2-yl)-N′-(3-methoxy-phenyl)(thio)ureas 11a,b were synthesized via amide coupling, starting from 6-methoxy-2ABT and 3-methoxyphenylisocyanate or 3-methoxyphenylisothiocyanate, respectively, Scheme 6 [104]. Boron tribromide was used for the methoxy group cleavage to yield 6-hydroxy,N-m-hydroxyphenyl-T(U)BTs 12a,b. The extension of the bridge to three units resulted in the inactive urea 12a, but interestingly, the thiourea 12b showed a moderate inhibitory activity on human 17β-HSD1 cancer cell line.
The N-(5-Benzyloxy, 7-Bromo-BT-2-yl)-N′-(ethyl)urea 13 was obtained in 75% yield from the reaction of 5-BnO,7-Br-2ABT with ethylisocyanate, Scheme 7 [105]. The pyridine ring was coupled to the C-7 of compound 13 to afford compound 14. Compound 14 was treated with methane sulfonic acid to yield the alcohol 15, which was converted to the triflate 16, and a Miyaura-borylation yielded the intermediate 17. Suzuki cross-coupling of boronic acid 17 with the corresponding 2-substituted-5-bromo-pyrimidine A, followed by saponification, yielded the target products 18ad. All these compounds showed bacterial DNA gyrase and topoisomerase IV inhibition. Antibacterial properties against DNA gyrase ATPase and potent activity against Staphylococcus aureus, Enterococcus faecalis, Streptococcus pyogenes and Haemophilus influenzae were tested. To increase drug-likeness analogues with a α-substituent to the carboxylic acid group, compounds 19ah and 20ad were designed and synthesized by using the same procedures. Compounds 19a and 19b had IC50 values of 0.012 and 0.008 μg/mL for S. aureus topoisomerase IV, comparable to their S. aureus DNA gyrase ATPase IC50 values. Additionally, compounds 19a and 19b showed specificity for bacterial topoisomerases, with no inhibition of human topoisomerase II.
The same procedure was used to synthesize a series of dual-targeting, alcohol- and diol-containing N-(5-pyrimidin-5-yl-7-pyridie-2-yl-BT-2-yl)-N′-(ethyl)ureas 21, 22, and 23, which showed superior antibacterial activity and drug-like properties, Figure 2 [106]. To improve the pharmacokinetic profile, the N-ethyl-UBTs 21ac were synthesized. The SAR study of this series showed that alcohol-containing compound had potent antibacterial activity against a primary panel of pathogenic bacteria compared to the carboxylic acids 19, Scheme 7. These compounds displayed potent antibacterial activity with minimum inhibitory concentration (MIC) against S. aureus 29,213 (0.03–0.06 μg/mL), S. pyogenes 0.06–0.12 μg/mL), and H. influenza 49,247a 0.25–1 μg/mL). Additionally, 21ac displayed a significative inhibitory activity against purified S. aureus T173 GyrB (IC50 0.25 μg/mL all cases) and S. pyogenes ParE (IC50 to 0.5–1 μg/mL). Compounds 21a,dm showed potency in the nanomolar range against DNA gyrase ATPase in a malachite green assay. Bulky groups at the tertiary, pseudobenzylic position of the pyrimidine ring resulted in a loss of activity, especially against the ParE mutant strain of S. pyogenes (21d diethyl, IC50 8 μg/mL; 21f cyclohexyl ring system, IC50 > 16 μg/mL). However, increased on-target ATPase inhibition was observed when a heteroatom is present into the alicyclic 6-membered ring (21gj), IC50s S. aureus GryB 0.25–8 μg/mL; S. pyogenes ParE 1–8 μg/mL, suggesting a conformational role, possibly a flattening of the ring. Reduced antibacterial activity against S. aureus 29213 (MIC > 16 μg/mL), S. pyogenes 51339 (MIC 8 μg/mL; IC50 > 16 μg/mL) and H. influenza 49247a (MIC 16 μg/mL) was observed when the ring was an azetidine (21l). However, strong potency (MIC 0.06–0.5 μg/mL) across the entire tested bacterial strains (with IC50s values of 0.5 and 2 μg/mL against S. aureus GryB and S. pyogenes ParE, respectively) was observed for the oxetane analogue 21m. Longer-chain alkyl R1 and small cycloalkyl groups (21nq) showed no effect on the potency of the serie toward the enzyme. However, the bulky t-butyl moiety (21p) gave a considerable drop in activity against Haemophilus influenza and against S. aereus in the presence of serum. The morpholine ring (21s) and diol (21r), designed to increase hydrophilicity, were well tolerated. Prompted by the possible steric constraint around the pyrimidine ring, the secondary alcohols 22ak and diol-containing molecules 23af (Figure 2) were explored to increase the solubility. Compounds 22af, with increased hydrophilicity and reduced protein binding, showed potent Gram-(+) antibacterial activity. Compounds 22ac increased the unbound fraction from 9% to 19%. However, this highlight was offset by an increase in MIC. Compounds without azetidine moieties 22ik increased this property. Compounds 23af, with small groups, did not significantly hinder the antibacterial activity. However, diol 23d showed some intolerance of larger rings.
The compound (Z)-ethyl-2-((Z)-2-(BT-2-ylimino)-4-oxo-3-phenylthiazolidin-5-ylidene)acetate 24 was designed to be synthesized under catalyst-free conditions, Scheme 8 [107]. Compound 24 was shown to be a receptor based on an internal charge transfer mechanism with the BT unit. The density functional theory calculations (DFT) complemented these results. The fluorescent emission of compound 24 in CH3OH/H2O (50:50 v/v) was quenched in the presence of Cu2+ and Hg2+, but not in the presence of other tested metal ions. Compound 24 was complexed in a 1:1 stoichiometry with Cu2+ and Hg2+ with detection limits of 0.36 and 2.49 mM, respectively. In addition, the interactions of 24 with aldose reductase inhibitor were investigated via the use of molecular docking studies.
N-(6-Bromo-BT-2-yl)-N′-(alkyl)ureas 25al, obtained from the reaction of 6-bromo-2ABT with CDI and alkylamine, were successively reacted with the sulfonamide compound 26, catalyzed by bis(pinacolato)diboron and PdCl2(dppf), to afford N-[6-(2,3-disubstituted-pyridin-5-yl)-BT-2-yl)-N′-(alkyl)ureas 27al and 28mr in 31–52% yield, Scheme 9 [108]. The antiproliferative activities of all synthesized compounds were tested in vitro against human colon HCT116, breast MCF-7, glioblastoma U87 MG, and adenocarcinoma A549 cell lines. The compounds with potent antiproliferative activity were tested for their acute oral toxicity and inhibitory activity against PI3Ks and mTORC1 pathways. The compound connected at 2-(dialkylamino)-N-ethylurea moiety at the 2-position of BT retained the antiproliferative and inhibitory activities against phosphatidylinositol-3-kinase (PI3K) and mamalian target of rapamicyn (mTOR). Additionally, their acute oral toxicity was reduced dramatically. Moreover, compound 27f inhibited tumor growth in a mice S180 homograft model. These results suggested that 1-(2-dialkyl-aminoethyl)-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)-UBTs could be used as potent PI3K inhibitors and anticancer agents with low toxicity. Secondly, compounds 27al showed potent antiproliferative activities against the four cancer cell lines, and the activities of most compounds 27 were near to that of positive controls. These data suggested that alkylurea moiety was tolerable at the 2-position of BT. It was deduced that the activity of compounds 27 is related to the substituent at the 2- and 3-positions of the pyridine ring as well as to the 1-position of urea moiety. The factor that compound 27f (0.3-0.45 μM) was more potent than compound 27b (0.71–5.26 μM), against four cancer lines, indicates that a methoxy group at the 2-position of pyridine ring in that compound may enhance the antiproliferative activity. Activities of compounds 27al were compared with that of compounds 28mq with different substituents at the 3-position of the pyridine ring, represented in Figure 3. Thirdly, the compounds containing the aryl-sulfonamino group at the 3-position of the pyridine ring, such as 4-methylphenylsulfonamino 27l (0.17–0.36 μM), exhibited better potent activity than those containing the cyclopropylsulfonamino group 28o (0.64–1.92 μM) and the cyano group 28n (2.30–8.38 μM) at the 3-position of the pyridine ring. If there was not a substituent at the 3-position of pyridine ring 28m (5.16–20 μM), the activity dramatically dropped. Fourthly, to investigate the SAR of compound 27, a range of selected amines were attached to the 1-position of urea moiety. The data indicate that the methyl urea 27d and cyclopropyl urea 27e showed a drop in their cell-based activity against the four cancer lines compared with the standard compound. However, 2-(N,N-disubstituted amino)-N-ethylureas 27fl displayed similar to enhanced antiproliferative activity against the four cancer lines compared with the positive controls. Fifthly, the replacement of BT core in compound 27g with thiazolo [5,4-b]pyridine moiety afforded compound 28r, which displayed an enhanced activity against HCT-116 (0.52 to 0.25 μM) and MCF-7 (0.75 to 0.26 μM) cells and a close activity against U87 MG (0.49 to 0.48 μM) and A549 (0.43 to 0.49 μM) cells. These results revealed that compound 28r is sensitive to PI3K mutant cells. In the cell-based activity, the IC50 values of compound 27f are near to that of the PI3K and mTOR dual inhibitor of the reference compound. In this sense, compound 27f was further investigated.
To compare the activities of compounds with different substituents at the 3-position of the pyridine ring, the N-[6-(5-substituted,6-methoxy-pyridin-3-yl)-BT-2-yl]-N′-[2-(morpholin-4-yl-)ethyl]urea 28mq were synthesized by using the same procedure, Figure 3.
Fifteen N-(BT-2-yl)-N′-(aryl)thioureas 29ag were synthesized from the corresponding arylisothiocianate, which underwent cyclization to 2-(N-thiazolidin-2-ene-4-one)-ABTs 30ag via treatment with chloroacetamide in acetonitrile, Scheme 10 [109]. All compounds were evaluated with respect to their cytotoxicity against human leukaemia/lymphoma and solid tumor-derived cell lines and of their antiviral activity against HIV-1 and representatives of ssRNA and dsDNA viruses. Compound 29e showed good activity against HIV-1 wild type and variants carrying clinically relevant mutations. A colorimetric enzyme immunoassay clarified its mode of action as a non-nucleoside inhibitor of the reverse transcriptase. The cyclization of compounds 29ag resulted in a whole decrease in their biological activities. Indeed, the anti-HIV activity of 29e is completely lost in the derivative 30e. Only 30b remains cytotoxic even at a lower value (CC50 from 1.6 to 9 μm). Compound 30b also seems to be less cytotoxic in the other antiproliferative assays, when compared with others and with its linear analogue 29b.
The 6-nitro-2ABT was reacted with ethylisocyanate to afford the N-(6-nitro-BT-2-yl)-N′-(Ethyl)urea 31, and the NO2 group was reduced to afford the N-(6-amino-BT-2-yl)-N′-(ethyl)urea 32, Scheme 11 [110]. Compound 32 was used to synthesize a series of N-(6-substituted-BT-2-yl)-N′-(ethyl)ureas 3337. Acylation of the 6-amino group of 32 with ethyl oxalyl chloride provided the ester 33a, which was hydrolyzed to the acid 33b. The compound 32 was coupled with the corresponding 2-(2-aminothiazol-4-yl)acetic acid derivative to yield carbonyl derivatives 34ac. Additionally, amine 35, produced by Boc-deprotection of 34b, was acylated with methyl malonyl chloride to produce 36a and with ethyl oxalyl chloride to produce 36b, which was hydrolized to the respective carboxylic acid 37. All compounds were evaluated for their Escherichia coli DNA gyrase inhibition. Among them, the UBTs 34ac and 36a were the most potent DNA gyrase inhibitors. Compound 34b showed an MIC of 50μM against an E. coli efflux pump-defective strain.
The treatment of 6-substituted-2ABTs with carbon disulfide in alkali media produced a dithiocarbamate as an intermediate, which was in situ reacted with dimethyl sulfate to afford 6-substituted-2-(S-methyl-dithiocarbamate)-BTs 38ad in excellent yields, Scheme 12 [111,112]. Compounds 38ad were reacted with ammonia or methyl anthranilate to be transformed to the N-(6-substituted-BT-2-yl)urea 39ad and the 2-[3-(6-substituted-BT-2-yl)-thioureido]benzoic acid methyl ester 41ad, respectively. The cyclization reaction of TBTs 39ad and 2-bromoacetophenone, in the presence of Et3N, affords N-(6-substitued-BT-2-yl)-4-phenyl-1,3-thiazol-2(3H)-imines 40ad. Additionally, S-methyl-dithiocarbamates 38ad reacted with methyl anthranilate to afford 3-(BT-2-yl)-2-thioxo-2,3-dihydroquinazolin-4(1H)-ones 42ad, which, after methylation with dimethyl sulfate, gave compound 43. Compounds 40ad were tested for anti-tumor activity against MCF-7, MAD-MD-231, HT-29, HeLa, Neuro-40a, K-562, and L-929 cell lines, and the MTT-assay revealed compound 40b with a phenolic segment, which resulted in having the best cytotoxicity (IC50 = 5.94 ± 1.98 μM) containing a phenolic segment. The apoptosis assay for compound 40b, carried out by flow cytometry, supported the results.

3. BT-Semi(Thio)Carbazide Derivatives

A set of 6-substituted-BT-thiosemicarbazones 4548 was synthesized from the corresponding thiosemicarbazides 44 to be screened for anticonvulsant and neurotoxic properties, Scheme 13 [113]. Majority of the compounds showed anticonvulsant activity against both maximal electroshock seizure (MES) and subcutaneous pentylenetetrazole (scPTZ) screens. Eight compounds showed good protection in the rat p.o. MES test at 30 mg kg−1. Compound 45a resulted as the most promising one with an ED50 of 17.86 and 6.07 mg kg−1 in mice i.p. and rat p.o., respectively. Compound 45a showed a weak ability to block the expression of fully kindled seizures. On the other hand, the 6-methyl BTyl-2-thiosemicarbazones 47ag had anticonvulsant activity in both mice i.p. and rat oral MES screen [114]. The 6-nitro-Btyl thiosemicarbazone 46a was the most promising one with anti-MES activity in mice i.p., rat i.p., and rat p.o. evaluations (MES screen 0.5 h; 300 mg kg−1) (scPTZ screen 4 h; 300 mg kg−1). All the compounds showed lesser or no neurotoxicity compared with the standard, phenytoin. The isatinimino derivatives exhibited better activity when compared with the benzylidene or acetophenone derivatives.
In 2007, a series of 6-substituted-BT-semicarbazones 49ao were prepared in satisfactory yield from the reaction of 6-substituted-BT-semicarbazides with p-substituted-acetophenones or -benzophenone, Scheme 14 [115]. All synthesized compounds were evaluated for their anticonvulsant, neurotoxicity, and other toxicity studies. Most compounds were active in maximal electroshock screen (MES). Compounds 49a, 49h, 49m, and 49o possessed 100% protection at both 0.5 h and 4 h time intervals, but for compound 49a, its percentage protection decreased to 83.3% at the 4 h interval, indicating the rapid onset but shorter duration of action under MES test conditions. The compounds containing more lipophilic substituents such as methyl and chloro groups on the BT ring were more active.
The BT-thiosemicarbazones 51ac in the presence of acetic anhydride were cyclized to produce the N-Acetyl, N-(5-aryl-4,5-dihydro-1,3,4-thiadiazol-2-yl)-2ABTs 52ac, Scheme 15 [116]. The in vitro activity against Schistosoma mansoni at three different dosage levels (10, 50, and 100 μg/mL) were screened for selected compound. The activity of BT-thiosemicarbazide 50 was proportional to its concentration, while the thiosemicarbazones 51ae showed a dramatic decrease in activity (51a,c were inactive). In addition, thiadiazole derivatives 52ac were moderately active. It was concluded that a dithiocarbamate link is required for better activity.
N-(6-Substituted-BT-2-yl)ureas 53, obtained from reaction of 6-substituted-2ABTs with HOCN, were functionalized with hydrazine, then reacted with substituted benzaldehydes to afford the corresponding BT-semicarbazone, and the cyclization of the BT-semicarbazones with chloroacetylchloride afforded a series of 6-substituted-BT(N-4-aryl-3-chloro-azetidinyl)semicarbazides 54at, Scheme 16 [117]. Their anticonvulsant, hepatotoxic and neurotoxic properties were evaluated. All compounds were screened for their anticonvulsant activity in an i.p. MES model and were compared with the standard drug phenytoin. Compounds 54f, 54n, and 54p exhibited 100% protection in the MES test. In the neurotoxicity and hepatotoxicity screening, all the compounds were devoid of toxicity at the dose of 30 mg/kg body weight. The study showed that the introduction of F or CH3 groups at the 6-position of BT moiety with H, OCH3 groups at the 3-position and OH, and OCH3 groups at the 4-position of the distant phenyl ring led to increased activity. The introduction of F, NO2, CH3, and OCH3 groups at the 6-position of the BT moiety and un-substituted distant phenyl ring showed a moderate decrease in activity. Compounds 54e, 54i, 54m, 54o, and 54q showed their ability to prevent seizure spread with 83% protection, whereas compounds 54a, 54b, 54d, 54l, and 54t showed 66% protection, indicating their ability to elevate the seizure threshold. Compounds 54c, 54g, 54h, 54j, 54k, 54r, and 54s showed 50% protection. Thus, most compounds displayed preferential MES selectivity. In the neurotoxicity screen, all the compounds were not toxic at the dose of 30 mg/kg body weight.
The N-(5-methyl-BT-2-yl)thiourea, obtained from the reaction of substituted 5-Me-2ABT with ammonium thiocyanate, was used as an intermediate to be reacted with hydrazine hydrate in ethylene glycol and then with substituted acetophenones to form the respective 5-methyl-BT-thiosemicarbazone 55ac, Scheme 17 [118]. All compounds were tested against antimicrobial activity by using Gram-(+) and Gram-(−) bacteria such as S. aureus, P. aeruginosa, and E. coli with ampicillin as standard compound. Derivatives showed a significant zone of inhibition against both Gram-(±) bacteria at 1 mg/mL. Then, compound 55a showed excellent activity against P. aeruginosa, while compound 55b showed excellent activity against E. coli, while compound 55c showed moderate activity against all three bacteria.
Six 6-substituted-BT-semicarbazides, obtained from the reaction of 6-substituted-2ABTs with sodium cyanate in acetic acid, then hydrazine in ethanol, were used as intermediates in the reaction with phenylisothiocyanate and concentrated sulfuric acid to afford N-(5-aminophenyl-[1,3,4]thiadiazol-2-yl)-2ABTs 56af, Scheme 18 [119]. The compounds were screened for anticancer activity (Human Chronic Myelogenous Leukemia) using in vitro assays. All compounds exhibited from a high to moderate anticancer activities. Compounds 56c and 56d showed the best anticancer potential because the presence of highly electronegative and electron-donating groups.
A series of N-(6-chloro-BT-2-yl)-N-([1,2,4]-triazolo-[3,4-b]-[1,3,4]-thiadiazole)amines 59ag and N-(6-chloro-BT-2-yl)-N-(5-substituted-[1,3,4]-oxadiazol-2-yl)amines 60ag and 61 were synthesized from the intermediate 6-chloro-BT-semicarbazide 57 obtained from 6-chloro-2ABT, NaOCN, and then hydrazine, Scheme 19 [120]. Their antimicrobial properties were investigated against one Gram-(+) bacteria (Staphylococcus aureus), three Gram-(−) bacteria (Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae), and five fungi (Penicillium citrinum, Candida albicans, Aspergillus flavus, Aspergi llus niger, and Monascus purpureus) using the serial plate dilution method. All the tested compounds exhibited moderate to good antibacterial and fungal inhibition at 12.5–100 μg/mL in DMSO. The triazolo-thiadiazole-ABTs 59ag were found to be more active than 1,3,4-oxadiazole-ABTs 60ag and 61 against all pathogenic bacterial and fungal strains.
6-chloro-BT-thiosemicarbazones 62ah were synthesized by Amir et al., Scheme 20 [121]. All synthesized compounds were screened against the in vivo anticonvulsant, and acute toxicity. Additionally, a 3D four-point pharmacophore measurements of the compounds was carried out to be compared with established anticonvulsants agents.
A series of 6-nitro-BT-semicarbazones 63au and 64 were designed and synthesized, to be evaluated as inhibitors of the rat brain MAO-B isoenzyme, Figure 4 [122]. Most compounds were potent inhibitors of MAO-B, with IC50 values from the nanomolar to micromolar range. Molecular docking studies with AutoDock 4.2 were performed to deduce the affinity and binding mode of these inhibitors toward the MAO-B active site. The free energies of binding (ΔG) and inhibition constants (Ki) of the docked compounds were calculated by the Lamarckian genetic algorithm (LGA) of Auto Dock 4.2. Good correlations were obtained between the calculated and experimental values. Compound 63d emerged as the lead MAO-B inhibitor, with top ranking in both the experimental MAO-B assay (IC50: 0.004 ± 0.001μΜ) and in computational docking studies (Ki: 1.08 μΜ). A binding mode analysis of potent inhibitors suggests that these compounds are well accommodated by the MAO-B active site through stable hydrophobic and hydrogen-bonding interactions. Interestingly, the 6-nitroBT moiety was found to be stabilized in the substrate cavity with the aryl or diaryl residues extending up into the entrance cavity of the active site. Thus, a binding site model consisting of three essential pharmacophoric features was proposed with the aim being for it to be used in the design of future MAO-B inhibitors.
On the other hand, the anticonvulsant activity of 2-amino-6-nitro-BT-semicarbazones 63 and 64 in various in vivo animal seizure models were investigated, Figure 4 [123]. MES, sscPTZ, and 6 Hz psychomotor seizure models were tested. Neurotoxicity was estimated by the rotarod test. Additionally, the compounds were assessed for their neuroprotective potential from excitotoxic insult using organotypic hippocampal slice culture neuroprotection assay. Various compounds exhibited excellent anticonvulsant activity in MES and scPTZ models compared to reference drugs, phenytoin and levetiracetam. The results of kainic acid (KA)-induced neuroprotection assay showed that compounds 63r and 63t were most potent with IC50 values of 101.00 ± 1.20 μM and 99.54 ± 1.27 μM, respectively. Both compounds attenuated KA-mediated cell death in organotypic hippocampal slice cultures. Some compounds were better antidepressants compared with the reference drug citalopram, when analyzed in a forced swim test. Since semicarbazones showed a profile resembling phenytoin, an intent was made to screen them against human neuronal sodium channel isoform (hNav1.2) by carrying out computational molecular docking using AutoDock 4.2. Compound 64c resulted in being the lead candidate with excellent in vivo MES activity and computationally better binding affinity than the reference drug phenytoin and also exhibited neuroprotection from excitotoxic insult in KA-induced neuroprotection assay (IC50 = 126.80 ± 1.24 μM). However, some of the active compounds were neurotoxic at their anticonvulsant doses. Optimization studies were proposed to reduce toxicity and develop them as novel epilepsy therapeutic agents.
The synthesis of potential anthelmintic agents N-(6-fluoro,7-chloro-BT-2-yl)-N′-(2-aryl, 4-one-thiazolidin-3-yl)ureas 68ae were achieved from reaction of the corresponding 6F,7Cl-2ABT and ethyl chloroformate to afford the ethyl 6F,7Cl-BT-yl-2-carbamate 65, which, via treatment with hydrazine hydrate in ethanol, gave the 6-fluro-7-chloro-BT-semicarbazide 66, Scheme 21 [124]. The BT-semicarbazide 66 was treated with aromatic aldehydes in absolute ethanol to yield the BT-semicarbazones 67ae. Thioglycolic acid and zinc chloride were added to the semicarbazones to afford thiazolidinone derivatives 68ae. All compounds showed good to moderate anthelmintic activity against earth worms Perituma posthuma compared with Albendazole as the standard drug. Compounds 68c and 68d showed good anthelmintic activity, whereas compounds 68a, 68b, and 68e displayed less or comparable anthelmintic activity.
The intermediates 6-substituted-BT-semicarbazides 69ae, obtained from reaction of 6-substituted-2ABT with phenyl chloroformate, then a hydrazinolysis, were reacted with substituted o-hydroxybenzaldehydes to afford a series of 6-substituted-BT-semicazone derivatives 7072, Scheme 22 [125]. These synthesized compounds were screened in vitro with respect to their cytotoxic activities against five cancer cell lines (NCI-H226, SK-N-SH, HT29, MKN45, and MDA-MB-231). Most of them exhibited moderate to excellent activity against the five cell lines. Compound 71g (procaspase-3 EC50 1.42 μM) and 72b (procaspase-3 EC50 0.25 μM) showed excellent antitumor activity with IC50 values from 0.14 μM to 0.98 μM against all cancer cell lines, which were 1.8–8.7 times more active than the first procaspase-activating compound (PAC-1) (procaspase-3 EC50 4.08 μM). The SAR analyses showed that a lipophilic group (benzyloxy or heteroaryloxy groups) at the 4-position of the 2-hydroxy phenyl ring, was beneficial to antitumor activity, and R1-substituents with nitrogen, which are positively charged at physiological pH, could improve antitumor activity. Additionally, it was confirmed that the steric effect of the 4-position substituent of the benzyloxy group had an influence on cytotoxicity.
On the other hand, the same procedure was used by Ma et al. to synthesize a different series of 6-substituted-BT-semicarbazones 7375 for antiproliferative activities and procaspase-3 kinase activities against five cell lines (MDA-MB-231, MNK-45, NCI-H226, HT-29, and SK-N-SH), Figure 5 [126]. The BT-semicarbazone 74e showed inhibitory activities against the five cell lines with IC50 and EC50 values from 0.4 to 0.92 μM and 0.31 μM, respectively. The SAR studies showed that the in vitro pharmacological activities of BT scaffold 74e are due to the introduction of phenyl and benzyloxyl substitutions.
From the condensation reaction of the respective 6-substituted-BT-semicarbazide with the corresponding aldehyde or ketone derived from substituted indole derivatives, a series of 6-substituted-BT-semicarbazones with an indole-based moiety 7678 were designed to be synthesized and screened for in vitro antitumor activity against four cancer cell lines (HT29, H460, A549, and MDA-MB-231), Figure 6 [127]. Most of them showed moderate to excellent activity against the four cell lines. The compounds 78azi exhibited better selectivity against the HT29 cancer cell line. Among these derivatives, the carboxamides 78za, showed potent antitumor activity with IC50 values of 0.015 μM for HT29, 0.28 μM for H460, 1.53 μM for A549, and 0.68 μM for MDA-MB-231. On the other hand, compound 78f exhibited excellent antitumor activity with IC50 values of 0.024, 0.29, 0.84, and 0.88 μM against the four cancer cell lines, respectively. The SAR studies showed that compound 78d exhibited the highest antitumor activity due to the presence of electron-withdrawing groups in the 4-position of the benzyl ring. In another work, it was found that compounds with fluoro-substituted benzyl-1H-indole moiety displayed more potent activity than those with phenyl moiety as compounds 78 (R = F), Figure 6 [128]. Compound 76c, the most promising, exhibited excellent antitumor activity (IC50 values of 0.015, 0.28, 1.53, and 0.68 μmol/L) against the four cell lines, respectively. Mechanism studies indicated that the marked pharmacological activity of compound 76c might be ascribed to activation of procaspase-3 (apoptosis-inducing) and cell cycle arrest, which was proposed as a lead for further structural modifications. Furthermore, a 3D-QSAR model (training set: q2 = 0.850, r2 = 0.987, test set: r2 = 0.811) was built to provide a comprehensive guide for further structural modification and optimization.
A series of seven 6-chloro-BT-semicarbazones 79ag were synthesized by reacting the 6-chloro-BT-semicarbazide with aryl aldehydes in refluxing ethanol. These compounds were condensed with thioglycolic acid to suffer ring closure to give compounds 80ag, Scheme 23 [129]. On the other hand, compounds 81ag were synthesized by reacting compounds 79ag with chloroacetyl chloride. All compounds were tested against one Gram-(+) bacteria (Staphylococcus aureus), three Gram-(−) bacteria (Pseudomonas aeruginosa, Klebsiella pneumonia, and Escherichia coli), and five fungi (Aspergillus niger, Aspergillus flavus, Candida albicans, Penicillium citrinum, and Monascus purpureus) using the serial plate dilution method. All the synthesized compounds showed from moderate to good antibacterial and antifungal inhibition (12.5–200 μg/mL in DMSO) compared with the standard drug Ofloxacin. The azetidin-2-ones 81ag were more active than thiazolidin-4-ones 80ag against all pathogenic bacterial and fungal strains. On the other hand, the in vivo anticonvulsant screening of these compounds revealed that 80c, 80g, and 80c had promising anticonvulsant activities without any neurotoxicity [130]. The evaluation of selected compounds for their in vitro GABA AT inhibition showed that compound 80c (IC50 15.26 μM) exhibited excellent activity compared with the vigabatrin as standard drug (IC50 39.72 μM), suggesting the potential of these BT derivatives as new anticonvulsants.
A series of 6-substituted-BT-semicarbazone compounds 82al was synthesized from the reaction of 6-substituted-BT-semicarbazide with the corresponding aldehyde or ketone for their in vitro evaluation in antiproliferative activity against procaspase-3 over-expression in cancer cell line U937 and a procaspase-3 no-expression in cancer cell line MCF-7, to rule out off-target effects, Scheme 24 [131]. Biological evaluation identified a series of BTs bearing a pyridine-semicarbazone moiety, 82j and 82k, with promising antiproliferative activity and significant selectivity. Mechanism studies showed that compounds 82j and 82k could induce apoptosis of cancer cells by activating procaspase-3 to caspase-3, and compound 82k showed the strongest procaspase-3 activation activity. SAR studies revealed that the presence of BT and an N,N,O-donor set is crucial for the antiproliferative activity and selectivity, and reducing the electron density of the N,N,O-donor set resulted in a dramatic decline in both. Furthermore, toxicity evaluation (zebrafish) in vivo and metabolic stability studies (human, rat, and mouse liver microsomes) were achieved to provide reliable guidance for further PK/PD studies.
Twelve 6/4-substituted-BT-semicarbazone-bis-sulfoxides 84al, as potential antibacterial agents, were designed and synthesized from the reaction of 6/4-substituted-BT-semicarbazides with carbon disulfide and then with the corresponding alkyl iodide to afford the intermediate dithioalkylcarboimidate derivative 83al, which were further oxidized in the presence of sodium tungstate dihydrate, Scheme 25 [132]. The results of the bioactivity assays showed that many compounds had significant in vitro inhibitory effects against Xanthomonas oryzae pv. oryzae (Xoo) and Xanthomonas citri pv. citri (Xac). Compound 84g had the best in vitro antibacterial activity against Xoo at a half-maximal effective concentration value of 11.4 μg/mL, which was superior to those of thiadiazole copper (TDC) and bismerthiazol (BMT). Compared with TDC and BMT, compound 84g was more effective in in vivo controlling rice bacterial leaf blight with curative and protection activities of 42.5% and 40.3%, respectively. Additionally, compound 84g influenced biofilm formation, inhibiting extracellular polysaccharide production, and ultimately, it reduced the pathogenicity of Xoo. All the results showed that bis-sulfoxides bearing acylhydrazone and BT moieties could be used as a base for the development of small-molecule pesticides with good antibacterial activity.
A series of N-(BT-2-yl)-N′-(substituted)(thio)ureas 8689 was obtained to be tested for their in vitro cytotoxicity against MCF-7 breast cancer cells, Scheme 26 [89]. The N-(BT-2-yl)-N′-(morpholin)urea 86a showed the highest cytotoxic activity. A docked pose of 86a bound to the G-quadruplex of the human telomere DNA active site using the Molecular Operating Environment (MOE) module was obtained. Moreover, all compounds were screened for their antimicrobial activity against Mycobacterium tuberculosis H37Rv, E. coli, S. aureus, and C. albicans. Compound 86a showed the best activity against M. tuberculosis H37Rv reducing the growth of MCF-7 cells to 76% at 10 μM, while other compounds were equipotent with ampicillin (12.5 and 25 μg mL−1) against S. aureus (88b) and E. coli (89a), respectively.

4. N-acyltUBT Derivatives

The treatment of 6/5-substituted-2ABTs with benzoyl isocyanates in acetone, afforded the N-(6/5-substituted-BT-2-yl)-N′-(benzoyl)thioureas 90ak, Scheme 27 [133]. All compounds were tested in vitro for their antimicrobial activity against Gram-(+) and Gram-(−) bacteria. They exhibited moderate to potent activity, as compared to the standard drugs. However, the compounds 90ak were less bioactive than the respective substituted-2ABTs. Compound 90a showed high activity against E. coli (35 mm of inhibition diameter at 2.0 mg/mL).
Rana et al. prepared a series of N-(6-substituted-BT-2-yl)-N′-(benzoyl)thioureas 91at in 61–88% yield from 6-substituted-2ABTs and substituted benzoyl isothiocyanates, Figure 7 [134]: All compounds were evaluated for their neurotoxicity, CNS depressant effects, anticonvulsant activity, and other toxicity studies. With exception of compounds 91m and 91qs, all compounds showed protection in the animal models of seizures. Compounds 91b, 91i, and 91p emerged as non-neurotoxic anticonvulsants and can be demanded to detect compounds possessing activities against generalized tonic-clonic (grand mal) and generalized absence (petit mal) seizures, respectively. None of the compounds showed neurotoxicity or liver toxicity. In a CNS depressant study, most compounds decreased the immobility time. For CNS depressant and anticonvulsant activity, the dose used was 30 mg/kg; however, the mechanism of action involved for both the activities, could not be proposed. However, further studies were suggested.
N-(BT-2-yl)-N′-(benzoyl)thioureas 90a and 92 were obtained from reaction of 2ABT with the corresponding in situ-generated benzoylthiocyanate, Figure 8 [135,136]. The N-benzoyl-TBTs structures were analyzed by X-ray diffraction analysis. The thiourea moiety was found to be in the thioamide form. The molecules of N-benzoyl-TBTs were found to be stabilized by the C—H---S and C—H---O intermolecular hydrogen bonds. Additionally, a pseudo-S(6) planar ring with an intramolecular N—H---O hydrogen bonding was formed with dihedral angles of 11.23 and 11.91 with the BT ring system and the phenyl ring, respectively.
On the other hand, Siddiqui et al. refluxed N-(6-substituted-BT-2-yl)-N′-(benzoyl)thioureas 93at in n-butanol to be cyclized to the phenyl-2H-[1,3,5]triazino[2,1-b][1,3]BT-2-thiones 94at, Scheme 28 [137]. All compounds were evaluated for their anticonvulsant, anti-nociceptive, hepatotoxic, and neurotoxic properties. Phenytoin was used as standard drug in the evaluation of their anticonvulsant activity in a mouse seizure model. Compounds 94a, 94c, 94f, and 94l showed total protection from 0.5 h to 4 h periods. None of the selected compounds showed any neurotoxicity or hepatotoxicity. Additionally, all compounds were tested for their anti-nociceptive activity by a thermal stimulus technique compared with diclofenac as the standard drug. Compounds 94o, 94q, and 94t resulted in having highly potent analgesic activity with p < 0.01.
Some N-(substituted-BT-2-yl)-N′-(purinoacetyl)thioureas 95aj were synthesized from the reaction of the in situ-generated 2-((2-amino-9-phenylsulfonyl)-9H-purin-6-yl)amino) acetyl isothiocyanate with substituted-2ABTs. Their in vitro antimicrobial activity against Gram-(+) and Gram-(−) strains and antifungal strains was evaluated using a micro dilution procedure, Scheme 29 [138]. All compounds proved to be effective (MIC values). Among them, compounds 95a (0.17 mM) and 95d (0.18 mM) exhibited very good activity against Kleb, and 95e (0.18 mM) exhibited very good activity against B. subtilis and A. niger. Compound 95e showed MIC value less than the standard drugs Ciprofloxacin (0.25 mM), Ampicillin (0.27 mM), and Fluconazole (0.35 mM). Electron-donating groups, such as halogen and methoxy groups, on the BT ring influenced the antibacterial and antifungal activity in comparison with the electron-withdrawing ones. Based on these results, it was proposed that the thiourea group connection between the BT and purine rings seems very important for the antimicrobial effect.
A series of twenty N-(5-substituted-BT-2-yl)-N′-acyl)thioureas 96 was efficiently synthesized and evaluated for antimicrobial and anti-proliferative activities against cancer cells, Scheme 30 [7]. These compounds exhibited a broad spectrum of activity against the tested microorganisms and showed higher activity against fungi than bacteria. Compounds 96ba, 96bb, 96bc, 96bd, and 96bd showed the greatest antimicrobial activity. Preliminary SAR studies showed that electronic factors in BT rings had a great effect on the antimicrobial activity. In preliminary MTT cytotoxicity studies, the compounds 96db, 96cb, and 96de resulted in being the most potent. In MCF-7 and HeLa cells, the range of the IC50 values was 18–26 μM and 38–46 μM, respectively.
A series of thirty N-(substituted-BT-2-yl)-N′-[2-methyl-4-oxoquinazolinethioyl-3(4H)]thioureas, 98100aj, was designed and synthesized. Their anticonvulsant activity in the seizure models scPTZ, 6-Hz, and MES were evaluated, Scheme 31 [139]. N-(substituted-BT-2-yl)-N′-(thiocarbamidoyl)ureas 97aj, obtained from reaction of UBTs with ammonium thiocyanate were reacted with 4-substituted-2-acetamidobenzoic acid to afford the compounds 98100aj. Compounds 100a (PI > 3.4; scPTZ) and 100f (PI > 26; MES > 4.2; scPTZ) exhibited comparable to higher activities with phenytoin (PI > 22; MES) and ethosuximide (PI > 3.0; scPTZ) as the reference drug. Additionally, they showed anticonvulsant activity against (S)-2-amino-3-(3-hydroxyl-5-methyl-4-isoxazolyl) propionic acid (AMPA)- and γ-amino butyric acid (GABA)-induced seizures. Compounds 100b and 100f could be provided to have activity at multiple receptors; thus, they were proposed as templates for future design, modification, and investigation to obtain more active analogs. However, further studies were proposed to precisely determine the mechanism and pharmacokinetic parameters.
N-(BT-2-yl)-N′-(benzoyl)thioureas 101ai, prepared from the reaction of 2ABT with the respective benzoyl isothiocyanate in refluxing acetone for 6 h, were converted in a gold(I)-mediated intramolecular trans-amidation via dethiocyanation to 2-(substituted-benzamide)-BTs 102ai, Scheme 32 [140]. The density functional theory calculations support the mechanism proposed in the transformation.

5. Guanidine Derivatives

Since 1968, TBTs have been used as intermediates to synthetize BT-2-yl-guanidines (GBTs); for instance, N-(substituted-BT-2-yl)-N′-(p-tolyl)thioureas 103ak, synthesized from reaction of substituted 2ABTs with p-tolylthiocyanate, were reacted with the corresponding ammonia, methylamine, or ethylamine in the presence of yellow lead oxide as the desulfurating agent to afford twenty-six N-(substituted-BT-2-yl)-N′-(p-tolyl)-N″-(alkyl)guanidines 104106, Scheme 33 [141]. Several of these were evaluated for their antibacterial and antitubercular activity against M. tuberculosis (H37Rv). All the tested compounds were inactive as antibacterials. However, compounds 104i and 105c were the most active as antituberculars.
On the other hand, several N-(substituted-BT-2-yl)-N′-(p-chlorophenyl)-N″-(alkyl)guanidines 107111 were synthesized in 50–90% yield, Figure 9. Compounds were screened for antitubercular and antibacterial activities [142]. The tested compounds were inactive in vitro at MIC = 80 μg/mL against M. tuberculosis (H37RV). All compounds were screened with respect to their antibacterial activity against S. aureus, B. subtilis, S. typhi, A. tumefaciens, and E. coli. Compounds 107f and 109f resulted in being the most active against B. subtilis and 107f, 107g, 108f, 109f, 109k, and 110i against S. aureus at 6.25 μg/mL. All compounds resulted in being inactive against A. tumefaciens, S. typhi, and E. coli at a maximum concentration of 100 μg/mL.
In 1971, the same author synthesized a series of N-(substituted-BT-2-yl)-N′-(benzyl)guanidines 112ak using the same procedure, Figure 10 [143].
In view of the antiprotozoal and antifungal activity of (substituted) benzothiazolyl guanidines, the same procedure was used to synthetize a series of twenty two N-p-bromophenyl-N′-(substituted)-benzothiazol-2-yl-N″-(n-propyl and n-butyl) guanidines 113a-k and 114a-k Figure 11 [144].
Four N-(BT-2-yl)-N′-(aryl)-N″-(amino)guanidines 115ad were synthesized from the amination-desulfhydration of N-(BT-2-yl)-N′-(aryl)thioureas, Scheme 34 [145]. These compounds were tested for their antimicrobial activity by the standard disc diffusion method. The activities were compared with that of standard strain of Escherichia coli, NCTC 10418. From the sensitive aminoguanidines, their MIC was further obtained.
N-(6-substituted-BT-2-yl)thioureas 116af, obtained from the reaction of 6-substituted-2ABTs with saturated ammoniumthiocynate, were treated with β-alanine in the presence of CuSO4.5H2O as the desulfurating agent to afford the N-(6-substituted-BT-2-yl)-N′-(2-carboxy-ethanoyl)guanidines 117af, Scheme 35 [146]: Compounds 117ae showed promising cytotoxicity against the human cervical cancer cell line (HeLa); however, compound 117f exhibited the most potent cytotoxicity. Compounds 116e and 117b were considered to be the most active antimicrobials, with a broad spectrum against both Gram-(+) and Gram-(−) bacteria.
Twelve acetylated N-(6-substituted-BT-2-yl)-N′-(sugar)-N″-(substituted)guanidines 119121 were obtained in 48–77% yield from the reaction of an alkyl or aryl amine with N-(6-substituted-BT-2-yl)-N′-(sugar)thioureas 118 in the presence of HgCl2, Scheme 36 [147]. The removal of protecting groups afforded guanidines 122124 in 76–95% yield. The 6-substituted-sugar-TBTs 118 were synthesized in 52–58% yield by the reaction of 6-substituted-2ABTs with the sugar thiocyanate derivative in anhydrous pyridine. Some of the synthesized guanidines displayed anti-influenza activity.
A series of N-(6-trifluoromethoxy-BT-2-yl)-N′-(alkyl)thioureas 125ad were synthetized from 6-trifluoromethoxy-2ABT with the corresponding alkylisothiocyanate. Among them, TBTs 125a and 125b were reacted with methyl iodide in acetone and then with gaseous ammonia to afford the respective guanidine 126a and 126b, Scheme 37 [148]. All compounds were tested by an in vitro protocol of ischemia/reperfusion injury, but only compounds 125ad showed a significant reduction in neuronal injury. The anti-oxidant properties of compounds 125ad were shown to be gifted with a direct ROS scavenging activity. Compounds 125b and 125d were tested for their activity on voltage-dependent Na+ and Ca2+ currents in neurons from rat piriform cortex. At 50 μM, compound 125b inhibited the transient Na+ current to a much smaller grade than rilusole, while 125d was near ineffective.

6. Conclusions

A bibliographic review about aspects of design, chemical synthesis, and biological activities related with (T)UBTs derivatives as potential therapeutic agents is herein presented.
Synthetic methodologies from 1968 to the present day, highlighting the approaches to transform (T)UBTs to afford derivative compounds with a variety of substituents, are illustrated with 37 Schemes and 11 Figures and are concluded with 148 references.
The work relates to recent reaction methodologies starting with (un)substituted-2ABT to afford the (T)UBTs, which were transformed into (1) (T)UBT derivatives, (2) BT-semi(thio)carbazide derivatives, (3) N-acyl-(T)UBT derivatives, and (4) GBT derivatives.
This topic provides information regarding the utility for medicinal chemists and the pharmaceutical industry, which are dedicated to the design, synthesis, and SAR studies of these interesting compounds. The main biological activities tested were included with the aim of provide a comprehensive overview of these compounds. The reader will find the required information to judge the suitability of compounds for testing other biological activities or pharmaceutical targets.
Most compounds were analyzed with respect to their biological activities such as ubiquitin ligase inhibitor and anti-helmintic, antitubercular, anti-nociceptive, anti-convulsant, anti-influenza, anti-inflammatory, anti-bacterial, anti-oxidant, anti-proliferative of cancer, antifungal, and antimicrobial activities. Some of these compounds are promising lead molecules to be developed as potent chemotherapeutic agents.
It was observed that the principal activities of the BT derivatives were anti-bacterial, anti-microbial, and anticancer activities.
The information provided in this review shows that (T)UBT derivatives have activity at multiple receptors. On this basis, these kinds of compounds can be proposed as templates for future design, modification, and investigation to synthesize more active analogs. On the other hand, researchers have proposed further studies to precisely determine the mechanism of action and pharmacokinetic parameters.

Author Contributions

Conceptualization, J.E.M.-W. and A.C.; formal Analysis, J.E.M.-W., M.C.R.-H. and E.V.G.-B.; investigation, A.C. and E.V.G.-B.; writing—original draft preparation, J.E.M.-W., A.C. and I.I.P.-M.; writing—review and editing, A.C., I.I.P.-M. and M.C.R.-H. All authors have read and agreed to the published version of the manuscript.

Funding

A. Cruz thanks the Secretaría de Investigación y Posgrado del Instituto Politécnico Nacional (SIP-IPN) for financial support, Grant no. 20230853-2023; M.C.R.-H. thanks CONACYT CB, Grant 319355-2022; A.C., J.E.M.-W. and M.C.R.-H. thank SIP-IPN for financial support, Grant 2140-2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Nomenclature

Description of Cancer cell lines mentioned in the text.
EKVX: Lung Adenocarcinoma; RPMI-8226: Roswell Park Memorial Institute 8226 Cells Plasma Cell Myeloma; OVCAR-4: Ovarian carcinoma; PC-3: Prostate cancer; CAKI-1: Human clear cell renal cell carcinoma; MDA-MB-435: Melanoma cells; T47D: Breast Invasive Ductal Carcinoma; B16-F10: Melanoma cells; U937: Acute Myeloid Leukemia cells; THP-1: Acute Myeloid Leukemia cells; HCT116: Colon Adenocarcinoma cells; MCF-7: Invasive Breast Carcinoma cells; U87 MG: Glioblastoma cells; A549: Lung Adenocarcinoma cells; MDA-MB-231: MD Anderson-Metastatic Breast-231 Cells; HT29: Colon Adenocarcinoma cells; HeLa: Cervical Adenocarcinoma cells; K-562: Chronic Myeloid Leukemia cells; NCI-H226: Pleural Mesothelioma, Epithelioid Type cells; SK-N-SH: Neuroblastoma cells; MKN-45: Diffuse Type Stomach Adenocarcinoma; L-929: NCTC clone 929 [L cell, L-929, derivative of Strain L] fibroblast cells; NCIH460 [H460]: Large Cell Lung Carcinoma cells; GSK-3β: Glycogen synthase kinase-3-betha; 17β-HSD1: 17β-hydroxysteroid dehydrogenase type 1 enzyme.

References

  1. Venkatachalam, T.K.; Mao, C.; Uckun, F.M. Effect of stereochemistry on the anti-HIV activity of chiral thiourea compounds. Bioorg. Med. Chem. 2004, 12, 4275–4284. [Google Scholar] [CrossRef] [PubMed]
  2. Lee, J.; Kang, S.-U.; Choi, H.-K.; Lee, J.; Lim, J.-O.; Kil, M.-J.; Jin, M.-K.; Kim, K.-P.; Sung, J.-H.; Chung, S.-J.; et al. Analysis of structure–activity relationships for the B-region of N-(3-acyloxy-2-benzylpropyl)-N0-[4-(methylsulfonylamino)-benzyl]thiourea analogues as vanilloid receptor antagonists: Discovery of an N-hydroxythiourea analogue with potent analgesic activity. Bioorg. Med. Chem. Lett. 2004, 14, 2291–2297. [Google Scholar] [CrossRef] [PubMed]
  3. Han, T.; Cho, J.H.; Oh, C.H. Synthesis and biological evaluation of 1β-methylcarbapenems having cyclic thiourea moieties and their related compounds. Eur. J. Med. Chem. 2006, 41, 825–832. [Google Scholar] [CrossRef]
  4. Khan, S.A.; Singh, N.; Saleem, K. Synthesis, characterization and in vitro antibacterial activity of thiourea and urea derivatives of steroids. Eur. J. Med. Chem. 2008, 43, 2272–2277. [Google Scholar] [CrossRef]
  5. Saeed, A.; Saeed, N.; Hummera, R.; Sadaf, R.; Hameed, A. Synthesis, characterization and antibacterial activity of some 1-aroyl-3-aryl tioureas. Chemistry 2009, 18, 152–158. [Google Scholar]
  6. Keche, A.P.; Hatnapure, G.D.; Tale, R.H.; Rodge, A.H.; Birajdar, S.S.; Kamble, V.M. A novel pyrimidine derivatives with aryl urea, thiourea and sulfonamide moieties: Synthesis, anti-inflammatory and antimicrobial evaluation. Bioorg. Med. Chem. Lett. 2012, 22, 3445–3448. [Google Scholar] [CrossRef] [PubMed]
  7. Saeed, S.; Rashid, N.; Jones, P.G.; Ali, M.; Hussain, R. Synthesis, characterization and biological evaluation of some thiourea derivatives bearing benzothiazole moiety as potential antimicrobial and anticancer agents. Eur. J. Med. Chem. 2010, 45, 1323–1331. [Google Scholar] [CrossRef]
  8. Pavić, K.; Rajić, Z.; Michnová, H.; Jampílek, J.; Perković, I.; Zorc, B. Second generation of primaquine ureas and bis-ureas as potential antimycobacterial agents. Mol. Divers. 2019, 23, 657–667. [Google Scholar] [CrossRef]
  9. Kaplancikli, Z.A.; Turan-Zitouni, G.; Ozdemir, A.; Revial, G. New triazole and triazolothiadiazine derivatives as possible antimicrobial agents. Eur. J. Med. Chem. 2008, 43, 155–159. [Google Scholar] [CrossRef]
  10. Koçyiğit-Kaymakçıoğlu, B.; Çalışır, M.M.; Özbek, B.; Ötük, G. Synthesis and antimicrobial activities of schiff bases derived from 4-amino-5-(1phenylethyl)-2,4-dihydro-3H-1,2,4-triazole-3-thione. Eur. J. Chem. 2010, 7, 458–464. [Google Scholar]
  11. Vega-Perez, J.M.; Perinan, I.; Argandona, M.; Vega-Holm, M.; Palo-Nieto, C.; Burgos-Morón, E.; López-Lázaro, M.; Vargas, C.; Nieto, J.J.; Iglesias-Guerra, F. Isoprenylthiourea and urea derivatives as new farnesyl diphos phate analogues: Synthesis and in vitro antimicrobial and cytotoxic activities. Eur. J. Med. Chem. 2012, 58, 591–612. [Google Scholar] [CrossRef] [PubMed]
  12. Karipcin, F.; Atis, M.; Sariboga, B.; Celik, H.; Tas, M. Structural, spectral, optical and antimicrobial properties of synthesized 1-benzoyl-3-furan2-ylmethyl-thiourea. J. Mol. Struct. 2013, 1048, 69–77. [Google Scholar] [CrossRef]
  13. Zhong, Z.; Xing, R.; Liu, S.; Wang, L.; Cai, S.; Li, P. Synthesis of acyl thiourea derivatives of chitosan and their antimicrobial activities in vitro. Carbohydr. Res. 2008, 343, 566–570. [Google Scholar] [CrossRef]
  14. Pingaew, R.; Sinthupoom, N.; Mandi, P.; Prachayasittikul, V.; Cherdtrakulkiat, R.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Synthesis, biological evaluation and in silico study of bis-thiourea derivatives as anticancer, antimalarial and antimicrobial agents. Med. Chem. Res. 2017, 26, 3136–3148. [Google Scholar] [CrossRef]
  15. Lee, J.; La, S.; Ahn, B.R.; Jeong, T.C.; Kim, D.H. Metabolism of 1-{3-[3-(4-cyanobenzyl)-3H-imidazol-4-yl]-propyl}-3-(6-methoxypyridin-3-yl)-1-(2-trifluoro-methyl-benzyl)tiourea (YH3945), a novel anticancer drug, in rats using the14C-labeled compound. Rapid Commun. Mass Spectrom. 2004, 18, 1901–1910. [Google Scholar] [CrossRef]
  16. Manjula, S.N.; Noolvi, N.M.; Parihar, K.V.; Reddy, S.A.M.; Ramani, V.; Gadad, A.K.; Singh, G.; Kutty, N.G.; Rao, C.M. Synthesis and antitumor activity of optically active thiourea and their 2-aminobenzo thiazole derivatives: A novel class of anticancer agents. Eur. J. Med. Chem. 2009, 44, 2923–2929. [Google Scholar] [CrossRef]
  17. Azimian, F.; Hamzeh-Mivehroud, M.; Mojarrad, J.S.; Hemmati, S.; Dastmalchi, S. Synthesis and biological evaluation of diaryl urea derivatives designed as potential anticarcinoma agents using de novo structure-based lead optimization approach. Eur. J. Med. Chem. 2020, 201, 112461. [Google Scholar] [CrossRef]
  18. Holla, B.S.; Veerenda, B.; Shivananda, K.; Boja, P. Synthesis characterization and anticancer activity studies on some Mannich bases derived from 1,2,4-triazoles. Eur. J. Med. Chem. 2003, 38, 759–767. [Google Scholar] [CrossRef]
  19. Ramadas, K.; Suresh, G.; Janarthanan, N.; Masilamani, S. Antifungal Activityof 1,3-DisubstitutedSymmetrical and Unsymmetrical Thioureas. Pestic. Sci. 1998, 52, 145–151. [Google Scholar] [CrossRef]
  20. Rodriguez-Fernandez, E.; Manzano, J.L.; Benito, J.J.; Hermosa, R.; Monte, E.; Criado, J.J. Thiourea, triazole and thiadiazine compounds and their metal complexes as antifungal agents. J. Inorg. Biochem. 2005, 99, 1558–1572. [Google Scholar] [CrossRef]
  21. Esteva-Font, C.; Phuan, P.-W.; Lee, S.; Su, T.; Anderson, M.O.; Verkman, A.S. Structure-activity analysis of thiourea analogs as inhibitors of UT-A and UT-B urea transporters. Biochim. Biophys. Acta 2015, 1848, 1075–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ravichandran, V.; Shalini, S.; Kumar, K.S.; Rajak, H.; Agrawal, R.K. Design, Synthesis and Evaluation of Thiourea Derivatives as Antimicrobial and Antiviral Agents. Lett. Drug Des. Discov. 2019, 16, 618–624. [Google Scholar] [CrossRef]
  23. Chen, M.-H.; Chen, Z.; Song, B.-A.; Bhadury, P.S.; Yang, S.; Cai, X.-J.; Hu, D.-Y.; Xue, W.; Zeng, S. Synthesis and Antiviral Activities of Chiral Thiourea Derivatives Containing an α-Aminophosphonate Moiety. J. Agric. Food Chem. 2009, 57, 1383–1388. [Google Scholar] [CrossRef] [PubMed]
  24. Karakus, S.; Koçyıgıt-Kaymakcioglu, B.; Toklu, H.Z.; Aricioglu, F.; Rollas, S. Synthesis and Anticonvulsant Activity of NewN-(Alkyl/Sub-stituted aryl)-N′-[4-(5-cyclohexylamino)-1,3,4-thiadiazole-2-yl)phenyl]tioureas. Arch. Pharm. Chem. Life Sci. 2009, 342, 48–53. [Google Scholar] [CrossRef]
  25. Mehrotra, S.; Roychowdhury, P.; Pandey, K.K.; Srivastava, P.K. Synthesis of 1-aryl-2-mercapto-4-aryl-1,6-dihydro-1,3,5-triazine-6-thione and their latentiation products as antithyroidal agent. Arch. Pharmacal Res. 1995, 18, 356. [Google Scholar] [CrossRef]
  26. Li, J.-Z.; Xue, S.-J.; Guo, Y.-L. Synthesis and Biological Activity of Oxidized Nicotinicacyl Thiourea Derivatives. Chin. J. Org. Chem. 2007, 27, 1278–1281. [Google Scholar]
  27. Moneer, A.A.; Mohammed, K.O.; El-Nassan, H.B. Synthesis of Novel Substituted Thiourea andBenzimidazole Derivatives Containing a PyrazoloneRing as Anti-Inflammatory Agents. Chem. Biol. Drug Des. 2016, 87, 784–793. [Google Scholar] [CrossRef]
  28. Mumtaz, A.; Majeed, A.; Zaib, S.; Rahman, S.U.; Hameed, S.; Saeed, A.; Rafique, H.; Mughal, E.; Maalik, A.; Hussain, I.; et al. Investigation of potent inhibitors of cholinesterase based on thiourea and pyrazoline derivatives: Synthesis, inhibition assay and molecular modeling studies. Bioorg. Chem. 2019, 90, 103036. [Google Scholar] [CrossRef]
  29. Begum, S.; Choudhary, M.I.; And Khan, K.M. Synthesis, phytotoxic, cytotoxic, acetylcholinesterase and butrylcholinesterase activities of N, N′-diaryl unsymmetrically substituted tioureas. Nat. Prod. Res. 2009, 23, 1719–1730. [Google Scholar] [CrossRef]
  30. Dixit, P.P.; Patil, V.J.; Nair, P.S.; Jain, S.; Sinha, N.; Arora, S.K. Synthesis of 1-[3-(4-benzotriazol-1/2-yl-3-fluoro-phenyl)-2-oxo-oxazolidin-5-ylmethyl]-3-substituted-thiourea derivatives as antituberculosis agents. Eur. J. Med. Chem. 2006, 41, 423–428. [Google Scholar] [CrossRef]
  31. Liav, A.; Angala, S.K.; Brennan, P.J.; Jackson, M. N-α-AldopentofuranosylN′-[p-(isoamyloxy)phenyl]-thiourea derivatives: Potential anti-TB therapeutic agents. Bioorg. Med. Chem. Lett. 2008, 18, 2649–2651. [Google Scholar] [CrossRef] [PubMed]
  32. Ekoue-Kovi, K.; Yearick, K.; Iwaniuk, D.P.; Natarajan, J.K.; Alumasa, J.; de Dios, A.C.; Roepe, P.D.; Wolf, C. Synthesis and antimalarial activity of new 4-amino-7-chloroquinolyl amides, sulfonamides, ureas and tioureas. Bioorg. Med. Chem. 2009, 17, 270–283. [Google Scholar] [CrossRef] [Green Version]
  33. Sunduru, N.; Srivastava, K.; Rajakumar, S.; Puri, S.K.; Saxena, J.K.; Chauhan, P.M.S. Synthesis of novel thiourea, thiazolidinedione, and thioparabanic acid derivatives of 4-aminoquinoline as potent antimalarials. Bioorg. Med. Chem. Lett. 2009, 19, 2570–2573. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, H.; Zhang, Y.; Wu, G.; Zhou, J.; Huang, W.; Hu, X. Synthesis and biological evaluation of sulfonylurea and thiourea derivatives substituted with benzenesulfonamide groups as potential hypoglycemic agents. Bioorg. Med. Chem. Lett. 2009, 19, 1740–1744. [Google Scholar] [CrossRef]
  35. Zhao, F.; Xiao, J.H.; Wang, Y.; Li, S. Synthesis of thiourea derivatives as CCR4 antagonists. Chin. Chem. Lett. 2009, 20, 296–299. [Google Scholar] [CrossRef]
  36. Esteves-Souza, A.; Pissinate, K.; Nascimento, M.G.; Grynberg, N.F.; Echevarria, A. Synthesis, cytotoxicity, and DNA-topoisomerase inhibitory activity of new asymmetric ureas and tioureas. Bioorg. Med. Chem. 2006, 14, 492–499. [Google Scholar] [CrossRef] [PubMed]
  37. Harriet, M.B.; Bret, F.; Paul, B. Riluzole. A review of its pharmacodynamics and Pharmacokinetic Properties and Therapeutic Potential in Amyotrophic Lateral Sclerosis. Drugs 1996, 52, 549–563. [Google Scholar]
  38. Pittenger, C.; Coric, V.; Banasr, M.; Bloch, M.; Krystal, J.H.; Sanacora, G. Rluzole in the Treatment of Mood and Anxiety Disorders. CNS Drugs 2008, 22, 761–786. [Google Scholar] [CrossRef]
  39. Gupta, A.; Rawat, S. Synthesis and Cyclization of Benzothiazole: Review. J. Curr. Pharm. Res. 2010, 3, 13–23. [Google Scholar]
  40. Rana, A.; Siddiqui, N.S.; Khan, S.A. Benzothiazoles: A new profile of biological activities. Indian J. Pharm. Sci. 2007, 69, 10–17. [Google Scholar]
  41. Chaudhar, P.; Sharma, P.K.; Sharma, A.; Varshney, J. Recent Advances in Pharmacological Activity of Benzothiazole Derivatives. Int. J. Curr. Pharm. Res. 2010, 2, 5–11. [Google Scholar]
  42. Akhilesh, G.; Swati, R. Therapeutic Importance of Benzothiazole: Review. Asian J. Res. Chem. 2010, 3, 821–836. [Google Scholar]
  43. Yadav, P.S.; Devprakash, D.; Senthilkumar, G.P. Benzothiazole: Different Methods of Synthesis and Diverse Biological Activities. Int. J. Pharm. Sci. Drug Res. 2011, 3, 01–07. [Google Scholar]
  44. Victor, F.; Raisa, R.d.R.; Claudia, R.B.G.; Thatyana, R.A.V. Chemistry and Biological Activities of 1,3-Benzothiazoles. Mini-Rev. Org. Chem. 2012, 9, 44–53. [Google Scholar]
  45. Jaiswal, S.; Mishra, A.P.; Srivastava, A. The Different Kinds of Reaction Involved in Synthesis of 2-Substituted Benzothiazole and its Derivatives: A Review. Res. J. Pharm. Biol. Chem. Sci. 2012, 3, 631–641. [Google Scholar]
  46. Ali, R.; Siddiqui, N. Biological Aspects of Emerging Benzothiazoles: A Short Review. J. Chem. 2013, 2013, 345198. [Google Scholar] [CrossRef]
  47. Henary, M.; Paranjpe, S.; Owens, E.A. Substituted benzothiazoles: Synthesis and medicinal characteristics. Heterocycl. Commun. 2013, 19, 89–99. [Google Scholar] [CrossRef]
  48. Sharma, P.C.; Sinhmar, A.; Sharma, A.; Rajak, H.; Pathak, D.P. Medicinal significance of benzothiazole scaffold: An insight view. J. Enzyme Inhib. Med. Chem. 2013, 28, 240–266. [Google Scholar] [CrossRef]
  49. Sompalle, R.; Roopan, S.M. Review on Benzothiazoles: Synthesis and Diverse Biological Activities. Chem. Sci. Rev. Lett. 2014, 2, 408–414. [Google Scholar]
  50. Xie, X.; Yan, Y.; Zhu, N.; Liu, G. Benzothiazoles exhibit broad-spectrum antitumor activity: Their potency, structure-activity and structure-metabolism relationship. Eur. J. Med. Chem. 2014, 76, 67–78. [Google Scholar] [CrossRef]
  51. Kamal, A.; Syed, M.-A.H.; Mohammed, S.M. Therapeutic potential of benzothiazoles: A patent review (2010–2014). Expert Opin. Ther. Pat. 2015, 25, 335–349. [Google Scholar] [CrossRef] [PubMed]
  52. Raju, G.N.; Karumudi, B.S.; Rao, N.R. Benzothiazole-Versatile heterocyclic nucleus in medicinal chemistry: A Review. Int. J. Pharm. Chem. 2015, 5, 104–114. [Google Scholar]
  53. Keri, R.S.; Patil, M.R.; Patil, S.A.; Budagumpi, S. A comprehensive review in current developments of benzothiazole-based molecules in medicinal chemistry. Eur. J. Med. Chem. 2015, 89, 207–251. [Google Scholar] [CrossRef] [PubMed]
  54. Seth, S. A Comprehensive Review on Recent advances in Synthesis & Pharmacotherapeutic potential of Benzothiazoles. Antiinflamm. Antiallergy Agents Med. Chem. 2015, 14, 98–112. [Google Scholar]
  55. Gill, R.K.; Rawal, R.K.; Bariwal, J. Recent advances in the chemistry and biology of benzothiazoles. Arch. Pharm. 2015, 348, 155–178. [Google Scholar] [CrossRef]
  56. Mene, D.; Kale, M. Exploration of Different Methodologies for Synthesizing Biologically Important Benzothiazoles: An Overview. Curr. Org. Synth. 2016, 13, 41–57. [Google Scholar] [CrossRef] [Green Version]
  57. Achaiah, G.; Sridhar Goud, S.N.; Praveen Kumar, K.; Mayuri, P. Review on 2-substituted benzothiazoles: Diversity of synthetic methods and biological activities. Int. J. Pharm. Sci. Res. 2016, 7, 1375–1382. [Google Scholar]
  58. Gulati, S.; Wakode, S.; Kaur, A.; Anand, K. Synthesis, biological activity and recent advancement of benzothiazoles: A classical Review. World J. Pharm. Pharm. Sci. 2017, 6, 1842–1869. [Google Scholar]
  59. Agarwal, S.; Gandhi, D.; Kalal, P. Benzothiazole: A Versatile and Multitarged Pharmacofore in the field of Medicinal Cehemistry. Lett. Org. Chem. 2017, 14, 724–729. [Google Scholar] [CrossRef]
  60. Banerjee, S.; Payra, S.; Saha, A. A Review on Synthesis of Benzothiazole Derivatives. Curr. Organocatal. 2017, 4, 164–181. [Google Scholar] [CrossRef]
  61. Shaista, A.; Amrita, P. Benzothiazole—A Magic Molecule. IJPSR 2017, 8, 4909–4929. [Google Scholar]
  62. Srivastava, A.; Mishra, A.P.; Chandra, S.; Bajpai, A. Benzothiazole derivative: A review on its pharmacological importance towards synthesis of lead. Int. J. Pharm. Sci. Res. 2019, 10, 1553–1566. [Google Scholar]
  63. Tariq, S.; Kanboj, P.; Amir, M. Therapeutic advancement of benzothiazole derivatives in the last decennial period. Arch. Pharm. 2019, 352, 1800170. [Google Scholar] [CrossRef] [Green Version]
  64. Abrol, S.; Bodla, R.B.; Goswami, C. A Comprehensive Review on Benzothiazole Derivatives for Their Biological Activities. Int. J. Pharm. Sci. Res. 2019, 10, 3196–3209. [Google Scholar]
  65. Liu, X.; Dong, Z.-B. A Review on Domino Condensation/Cyclization Reactions for the Synthesis of 2-Substituted 1,3-Benzothiazole Derivatives. Eur. J. Org. Chem. 2020, 2020, 408–419. [Google Scholar] [CrossRef]
  66. Gao, X.; Liu, J.; Zuo, X.; Feng, X.; Gao, Y. Recent Advances in Synthesis of Benzothiazole Compounds Related to Green Chemistry. Molecules 2020, 25, 1675. [Google Scholar] [CrossRef] [Green Version]
  67. Pathak, N.; Rathi, E.; Kumar, N.; Kini, S.G.; Rao, C.M. A Review on Anticancer Potentials of Benzothiazole Derivatives. Mini-Rev. Med. Chem. 2020, 20, 12–23. [Google Scholar] [CrossRef]
  68. Mahesh, B.; Shiddappa, B.L. Structural Activity Relationship and Importance of Benzothiazole Derivatives in Medicinal Chemistry: A Comprehensive Review. Mini-Rev. Org. Chem. 2020, 17, 323–350. [Google Scholar]
  69. Elgemeire, G.H.; Azzam, R.A.; Osman, R.R. Recent advances in synthesis, metal complexes and biological evaluation of 2-aryl, 2-pyridyl and 2-pyrimidylbenzothiazoles as potential chemotherapeutics. Inorg. Chim. Acta 2020, 502, 119302. [Google Scholar] [CrossRef]
  70. Irfana, A.; Batoola, F.; Naqvia, S.A.Z.; Islamb, A.; Osmanc, S.M.; Nocentinid, A.; Alissae, S.A.; Supuran, C.T. Benzothiazole derivatives as anticancer agents. J. Enzyme Inhib. Med. Chem. 2020, 35, 265–279. [Google Scholar] [CrossRef] [Green Version]
  71. Sharma, P.C.; Sharma, D.; Sharma, A.; Bansal, K.K.; Rajak, H.; Sharma, S.; Thakur, V.K. New horizons in benzothiazole scaffold for cancer therapy: Advances in bioactivity, functionality, and chemistry. Appl. Mater. Today 2020, 20, 100783. [Google Scholar] [CrossRef]
  72. Sharma, P.C.; Bansal, K.K.; Sharma, A.; Sharma, D.; Deep, A. Thiazole-containing compounds as therapeutic targets for cancer therapy. Eur. J. Med. Chem. 2020, 188, 112016. [Google Scholar] [CrossRef] [PubMed]
  73. Elamin, M.B.; Elaziz, A.A.E.S.A.; Abdallah, E.M. Benzothiazole moieties and their derivatives as antimicrobial and antiviral agents: A mini-review. Int. J. Res. Pharm. Sci. 2020, 3, 3309–3315. [Google Scholar] [CrossRef]
  74. Law, C.S.W.; Yeong, K.Y. Current trends of benzothiazoles in drug discovery: A patent review (2015–2020). Expert Opin. Ther. Pat. 2022, 32, 299–315. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, J.; Liu, Y.; Zhang, Y.; Hu, L.; Han, S. Progress in Synthesis of 2-Substituted Benzothiazole Compounds. Chin. J. Org. Chem. 2021, 41, 1053–1071. [Google Scholar] [CrossRef]
  76. Dhadda, S.; Raigar, A.K.; Saini, K.; Manju, G.A. Benzothiazoles: From recent advances in green synthesis to anti-cancer potential. Sustain. Chem. Pharm. 2021, 24, 100521. [Google Scholar] [CrossRef]
  77. Sumit; Kumar, A.; Mishra, A.K. Advancement in Pharmacological Activities of Benzothiazole and its Derivatives: An Up to Date Review. Mini-Rev. Med. Chem. 2021, 21, 314–335. [Google Scholar] [CrossRef]
  78. Agarwal, D.K.; Sahiba, N.; Sethiya, A.; Soni, J.; Teli, P.; Agarwal, S.; Goyal, P.K. Insight View on Synthetic Strategies and Biological Applications of Pyrimidobenzothiazoles. Mini-Rev. Org. Chem. 2021, 18, 1012–1025. [Google Scholar] [CrossRef]
  79. Mohammad, A.; Mohd, I. A Mini-Review on Pharmacological Importance of Benzothiazole Scaffold. Mini-Rev. Org. Chem. 2021, 18, 1086–1097. [Google Scholar]
  80. Thube, U.S.; Pawar, P.Y.; Sawant, R.L. Synthesis and Antidiabetic Evaluation of Some 2-substituted Benzothiazole Derivatives. Eur. J. Mol. Clin. Med. 2021, 8, 2343–2350. [Google Scholar]
  81. Bhagdev, K.; Sarkar, S. Benzothiazole: As an Antidiabetic Agent. Ann. Rom. Soc. Cell Biol. 2021, 25, 20269–20285. [Google Scholar]
  82. Larisa, V.; Zhilitskaya, L.V.; Yarosh, N.O. Synthesis of biologically active derivatives of 2-aminobenzothiazole. Chem. Heterocycl. Comp. 2021, 57, 369–373. [Google Scholar]
  83. Bhoge, N.D.; Mohite, P.B.; Deshmukh, V.K.; Magare, B.K. A Comprehensive Review on Synthetic Strategy of Benzothiazole Lead and Pharmacological Importance. Indian J. Pharm. Drug Stud. 2021, 2, 15–19. [Google Scholar]
  84. García-Báez, E.V.; Padilla-Martínez, I.I.; Tamay-Cach, F.; Cruz, A. Benzothiazoles from Condensation of o-Aminothiophenoles with Carboxylic Acids and Their Derivatives: A Review. Molecules 2021, 26, 6518. [Google Scholar] [CrossRef] [PubMed]
  85. Haider, K.; Shrivstava, N.; Pathak, A.; Dewangan, R.P.; Yahya, S.; Yar, S. Recent advances and SAR study of 2-substituted benzothiazole scaffold based potent chemotherapeutic agents. Results Chem. 2022, 4, 100258. [Google Scholar] [CrossRef]
  86. Chanchal, S.; Rajnish, K.; Avijit, M.; Salahuddin, A.K.; Rakesh, S.; Shivali, M.; Mohd, A.M. Benzothiazole: Synthetic Strategies, Biological Potential, and Interactions with Targets. Mini-Rev. Org. Chem. 2022, 19, 242–256. [Google Scholar]
  87. Asiri, Y.I.; Alsayari, A.; Muhsinah, A.B.; Mabkhotc, Y.N.; Hassan, M.Z. Benzothiazoles as potential antiviral agents. J. Pharm. Pharmacol. 2020, 72, 1459–1480. [Google Scholar] [CrossRef] [PubMed]
  88. Bhagdev, K.; Sarkar, S. Benzothiazole: As an Antiviral Agent. Med. Sci. Forum 2021, 7, 9. [Google Scholar] [CrossRef]
  89. Abdel-Rahman, H.M.; Morsy, M.A. Novel benzothiazolyl urea and thiourea derivatives with potential cytotoxic and antimicrobial activities. J. Enzyme Inhib. Med. Chem. 2007, 22, 57–64. [Google Scholar] [CrossRef] [PubMed]
  90. Lind, P.T.; Morin, J.M., Jr.; Noreen, R.; Ternansky, R.J. Thiourea Derivatives and Methods for Inhibition of HIV and Related Viruses. WO Patent 9303022, 18 February 1993. [Google Scholar]
  91. Kumbhare, R.M.; Dadmal, T.; Kosurkara, U.; Sridhar, V.; Rao, J.V. Synthesis and cytotoxic evaluation of thiourea and N-bis-benzothiazolederivatives: A novel class of cytotoxic agents. Bioorg. Med. Chem. Lett. 2012, 22, 453–455. [Google Scholar] [CrossRef]
  92. Hartley, D.; Kidd, H. The Agrochemical Handbook; The Royal Society of Chemistry: Nottingham, UK, 1987. [Google Scholar]
  93. Wegler, R.; Eue, L. Chemie der Pflanzenschutz- und Schädlingsbekämp- fungsmittel. In Herbizide; Springer: Berlin, Germany, 1977; Volume 5. [Google Scholar]
  94. Ondrej, B.; Lukas, H.; Laura, A.; Rafael, D.; Patrick, G.; Marketa, B.; Ondrej, S.; Karel, M.; Kamil, K.; Terry, S.; et al. 6-Benzothiazolyl Ureas, Thioureas and Guanidines are Potent Inhibitors of ABAD/17&:β-HSD10 and Potential Drugs for Alzheimer’s Disease Treatment: Design, Synthesis and in vitro Evaluation. J. Med. Chem. 2017, 13, 345–358. [Google Scholar]
  95. Hroch, L.; Benek, O.; Guest, P.; Aitken, L.; Soukup, O.; Janockova, J.; Musil, K.; Dohnal, V.; Dolezal, R.; Kuca, K.; et al. Design, synthesis and in vitro evaluation of benzothiazole-based ureas as potential ABAD/17β-HSD10 modulators for Alzheimer’s disease treatment. Bioorg. Med. Chem. Lett. 2016, 26, 3675–3678. [Google Scholar] [CrossRef] [Green Version]
  96. Aitken, L.; Benek, O.; McKelvie, B.E.; Hughes, R.E.; Hroch, L.; Schmidt, M.; Major, L.L.; Vinklarova, L.; Kamil Kuca, K.; Smith, T.K.; et al. Novel Benzothiazole-Based Ureas as 17β-HSD10 Inhibitors, A Potential Alzheimer’s Disease Treatment. Molecules 2019, 24, 2757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Schmidt, M.; Benek, O.; Vinklarova, L.; Hrabinova, M.; Zemanova, L.; Chribek, M.; Kralova, V.; Hroch, L.; Dolezal, R.; Lycka, A.; et al. Benzothiazolyl Ureas are Low Micromolar and Uncompetitive Inhibitors of 17β-HSD10 with Implications to Alzheimer’s Disease Treatment. Int. J. Mol. Sci. 2020, 21, 2059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Lukas, H.; Patrick, G.; Ondrej, B.; Ondrej, S.; Jana, J.; Rafael, D.; Kamil, K.; Laura, A.; Terry, K.S.; Frank, G.-M.; et al. Synthesis and evaluation of frentizole-based indolyl thiourea analogues as MAO/ABAD inhibitors for Alzheimer’s disease treatment. Biorg. Med. Chem. 2017, 25, 1143–1152. [Google Scholar]
  99. Rosales-Hernández, M.C.; Mendieta-Wejebe, J.E.; Padilla-Martínez, I.I.; García-Báez, E.V.; Cruz, A. Synthesis and Biological Importance of 2-(thio)ureabenzothiazoles. Molecules 2022, 27, 6104. [Google Scholar] [CrossRef]
  100. Sutoris, V.; Algas, J. Benzothiazole compounds. III. Synthesis and biological activity of substituted N-(2-benzothiazolyl)ureas and benzothiazolyldihydrouracils. Chem. Zvesti (Chem. Pap.) 1973, 27, 829–833. [Google Scholar]
  101. Youssef, K.M.; Al-Abbdullah, E.; El-Khamees, H. Synthesis of sulofenur analoges as antitumour agents: Part II. Med. Chem. Res. 2002, 11, 481–503. [Google Scholar]
  102. Pareek, D.; Chaudhary, M.; Pareek, P.K.; Kantc, R.; Ojha, K.G.; Iraqi, S.M.U.; Pareek, A. Synthesis of some biologically important 2-thiobarbituric acid derivatives incorporating benzothiazole moiety. Der Pharm. Lett. 2010, 2, 274–283. [Google Scholar]
  103. LoMonte, F.; Kramer, T.; Boländer, A.; Plotkin, B.; Eldar-Finkelman, H.; Fuertes, A.; Dominguez, J.; Schmidt, B. Synthesis and biological evaluation of glycogen synthase kinase 3 (GSK-3) inhibitors: An fast and atom efficient access to 1-aryl-3-benzylureas. Bioorg. Med. Chem. Lett. 2011, 21, 5610–5615. [Google Scholar]
  104. Spadaro, A.; Negri, M.; Marchais-Oberwinkler, S.; Bey, N.; Frotscher, M. Hydroxybenzothiazoles as New Nonsteroidal Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1). PLoS ONE 2012, 7, e29252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Axford, L.C.; Anderson, K.H.; Andrau, L.N.; Atherall, J.; Barker, S.; Bennett, J.M.; Blair, M.; Collins, I.; Czaplewski, L.G.; Davies, D.T.; et al. Design, synthesis and biological evaluation of α-substituted isonipecotic acid benzothiazole analogues as potent bacterial type II topoisomerase inhibitors. Bioorg. Med. Chem. Lett. 2013, 23, 6598–6603. [Google Scholar] [CrossRef] [PubMed]
  106. Palmer, J.T.; Axford, L.C.; Barker, S.; Bennett, J.M.; Blair, M.; Collins, I.; Davies, D.T.; Ford, L.; Gannon, C.T.; Lancett, P.; et al. Discovery and in vivo evaluation of alcohol-containing benzothiazoles as potent dual-targeting bacterial DNA supercoiling inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, 4215–4222. [Google Scholar] [CrossRef] [PubMed]
  107. Wagh, Y.B.; Kuwar, A.; Sahoo, S.K.; Galluccic, J.; Dalal, D.S. Highly selective fluorimetric sensor for Cu2+ and Hg2+ using a benzothiazole-based receptor in semi-aqueous media and molecular docking studies. RSC Adv. 2015, 5, 45528–45534. [Google Scholar] [CrossRef]
  108. Xie, X.-X.; Li, H.; Wang, J.; Mao, S.; Xin, M.-H.; Lu, S.-M.; Zhang, S.-Q. Synthesis and anticancer effects evaluation of 1-alkyl-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)benzo[d]thiazol-2-yl)urea as anticancer agents with low toxicity. Bioorg. Med. Chem. 2015, 23, 6477–6485. [Google Scholar] [CrossRef]
  109. Bielenica, A.; Sanna, G.; Madeddu, S.; Struga, M.; Jóźwiak, M.; Anna, E.; Kozioł, A.E.; Sawczenko, A.; Materek, I.B.; Serra, A.; et al. New Thiourea and 1,3-thiazolidin-4-one Derivatives Effective on the HIV-1Virus. Chem. Biol. Drug Des. 2017, 90, 883–891. [Google Scholar] [CrossRef]
  110. Tomašič, T.; Barančoková, M.; Zidar, N.; Ilaš, J.; Tammela, P.; Kikelj, D. Design, synthesis, and biological evaluation of 1-ethyl-3-(thiazol-2-yl)urea derivatives as Escherichia coliDNA gyrase inhibitors. Arch. Pharm. Chem. Life Sci. 2018, 351, e1700333. [Google Scholar] [CrossRef] [Green Version]
  111. Eshkil, F.; Eshghi, H.; Saljooghi, A.S.; Bakavoli, M. Synthesis of New Benzothiazole Derivatives and Evaluation of Cytotoxicity of N-(6-Substitued-1,3-benzothiazol-2-yl)-4-phenyl-1,3-thiazol-2(3H)-imine Compounds. Org. Chem. Res. 2019, 5, 87–94. [Google Scholar]
  112. Eshkil, F.; Eshghi, H.; Saljooghi, A.S.; Bakavoli, M.; Rahimizadeh, M. Benzothiazole Thiourea Derivatives as Anticancer Agents: Design, Synthesis, and Biological Screening. Russ. J. Bioorg. Chem. 2017, 43, 576–582. [Google Scholar] [CrossRef]
  113. Yogeeswari, P.; Sriram, D.; Jit, L.R.J.S.; Sathiyanesan Sathish Kumar, S.S.; James, P.; Stables, J.P. Anticonvulsant and neurotoxicity evaluation of some 6-chlorobenzothiazolyl-2-thiosemicarbazones. Eur. J. Med. Chem. 2002, 37, 231–236. [Google Scholar] [CrossRef]
  114. Yogeeswari, P.; Sriram, D.; Mehta, S.; Nigam, D.; Kumar, M.M.; Murugesan, S.; Stables, J.P. Anticonvulsant and neurotoxicity evaluation of some 6-substituted benzothiazolyl-2-thiosemicarbazones. Il. Farm. 2005, 60, 1–5. [Google Scholar] [CrossRef] [PubMed]
  115. Siddiqui, N.; Rana, A.; Khan, S.A.; Bhat, M.A.; Haque, S.E. Synthesis of benzothiazole semicarbazones as novel anticonvulsants–the role of hydrophobic domain. Bioorg. Med. Chem. Lett. 2007, 17, 4178–4182. [Google Scholar] [CrossRef]
  116. Mahran, M.A.; William, S.; Ramzy, F.; Sembel, A.M. Synthesis and in vitro Evaluation of New Benzothiazole Derivatives as Schistosomicidal Agents. Molecules 2007, 12, 622–633. [Google Scholar] [CrossRef] [PubMed]
  117. Siddiqui, N.; Rana, A.; Khan, S.A.; Haque, S.E.; Alam, M.S.; Ahsan, W.; Arshad, M.F. Anticonvulsant and toxicity evaluation of newly synthesized 1- [2-(3,4-disubstituted phenyl)-3-chloro-4-oxoazetidin-1-yl]-3-(6-substituted-1,3-benzothiazol-2-yl)ureas. Acta Chim. Slov. 2009, 56, 462–469. [Google Scholar]
  118. Pandurangan, A.; Sharma, A.; Sharma, N.; Sharma, P.K.; Visht, S. Synthesis and structural studies of novel benzothiazole derivative and evaluation of their antimicrobial activity. Der Pharma Chem. 2010, 2, 316–324. [Google Scholar]
  119. Sekar, V.; Perumal, P.; Gandhimathi, S. Screening of anticancer activity in newly synthesized benzothiazole derivatives. J. Pharm. Sci. Res. 2011, 3, 1520–1524. [Google Scholar]
  120. Gilani, S.J.; Khan, S.A.; Siddiqui, N.; Verma, S.P.; Mullick, P.; Alam, O. Synthesis and in vitro antimicrobial activity of novel N-(6-chlorobenzo[d]thiazol-2-yl) hydrazine carboxamide derivatives of benzothiazole class. J. Enzyme Inhib. Med. Chem. 2011, 26, 332–340. [Google Scholar] [CrossRef]
  121. Amir, M.; Asif, S.; Ali, I.; Hassan, M.Z. Synthesis of benzothiazole derivatives having acetamido and carbothioamido pharmacophore as anticonvulsant agents. Med. Chem. Res. 2012, 21, 2661–2670. [Google Scholar] [CrossRef]
  122. Tripathi, R.K.P.; Goshain, O.; Ayyannan, S.R. Design, Synthesis, in vitro MAO-B Inhibitory Evaluation and Computational Studies of Some 6-Nitrobenzothiazole-Derived Semicarbazones. Chem. Med. Chem. 2013, 8, 462–474. [Google Scholar] [CrossRef]
  123. Tripathi, R.K.P.; Ayyannan, S.R. Anticonvulsant activity, organotypic hippocampal neuroprotection assay and in-silico sodium channel blocking potential of 2-amino-6-nitrobenzothiazole derived semicarbazones. Biomed. Pharmacother. 2017, 95, 1451–1460. [Google Scholar] [CrossRef]
  124. Sarkar, S.; Dwivedi, J.; Chauhan, R. Synthesis of 1-[2 (substituted phenyl)-4-oxothiazolidin-3-yl]-3-(6-fluro-7-chloro-1,3-benzothiazol-2-yl)-ureas as anthelmintic agent. J. Pharm. Res. 2013, 7, 439–442. [Google Scholar] [CrossRef]
  125. Chen, J.; Ma, D.; Lu, K.; Wang, L.; Han, X.; Zhao, Y.; Gong, P. Design, synthesis and structure activity relationships of novel benzothiazole derivatives bearing the ortho-hydroxy N-carbamoylhydrazone moiety as potent antitumor agents. Eur. J. Med. Chem. 2014, 86, 257–269. [Google Scholar]
  126. Zhang, J.; Ma, G.; Han, X.; Bao, G.; Wang, L.; Zhai, X.; Gong, P. Synthesis and biological evaluation of benzothiazole derivatives bearing the ortho-hydroxy-N-acylhydrazone moiety as potent antitumor agents. Arch. Pharm. Chem. Life Sci. 2014, 347, 936–949. [Google Scholar]
  127. Ma, J.; Bao, G.; Wang, L.; Li, W.; Xu, B.; Du, B.; Lv, J.; Zhai, X.; Gong, P. Design, synthesis, biological evaluation and preliminary mechanism study of novel benzothiazole derivatives bearing indole-based moiety as potent antitumor agents. Eur. J. Med. Chem. 2015, 96, 173–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ma, J.; Hu, G.; Xie, L.; Chen, L.; Xu, B.; Gong, P. Design, synthesis and biological evaluation of novel benzothiazole derivatives bearing semicarbazone moiety as antitumor agents. Chem. Res. Chin. Univ. 2015, 31, 958–963. [Google Scholar] [CrossRef]
  129. Gilani, S.J.; Nagarajan, K.; Dixit, S.P.; Taleuzzaman, M.; Khan, S.A. Benzothiazole incorporated thiazolidin-4-ones and azetidin-2-ones derivatives: Synthesis and in vitro antimicrobial evaluation. Arabian J. Chem. 2016, 9, S1523–S1531. [Google Scholar] [CrossRef] [Green Version]
  130. Gilani, S.J.; Hassan, M.Z.; Imam, S.S.; Kala, C.; Dixit, S.P. Novel benzothiazole hydrazine carboxamide hybrid scaffolds as potential in vitro GABA AT enzyme inhibitors: Synthesis, molecular docking and antiepileptic evaluation. Bioorg. Med. Chem. Lett. 2019, 29, 1825–1830. [Google Scholar] [CrossRef]
  131. Ma, J.; Ni, X.; Gao, Y.; Huang, K.; Liu, J.; Wang, Y.; Chen, R.; Wang, C. Identification and biological evaluation of novel benzothiazole derivatives bearing a pyridine-semicarbazone moiety as apoptosis inducers via activation of procaspase-3 to caspase-3. Med. Chem. Commun. 2019, 10, 465–477. [Google Scholar] [CrossRef]
  132. Zhang, J.; Wei, C.; Li, S.; Hu, D.; Song, B. Discovery of novel bis-sulfoxide derivatives bearing acylhydrazone and benzothiazole moieties as potential antibacterial agents. Pestic. Biochem. Physiol. 2020, 167, 104605. [Google Scholar] [CrossRef]
  133. Saeed, A.; Rafique, H.; Hameed, A.; Rasheed, S. Synthesis and Antibacterial Activity of Some New 1-aroyl-3-(substituted-2-benzothiazolyl)tioureas. Pharm. Chem. J. 2008, 42, 191–195. [Google Scholar] [CrossRef]
  134. Rana, A.; Siddiqui, N.; Khan, S.A.; Ehtaishamul, H.S.; Bhat, M.A. N-{[(6-substituted-1,3-benzothiazole-2-yl)amino]carbonothioyl}-2/4-substituted benzamides: Synthesis and pharmacological evaluation. Eur. J. Med. Chem. 2008, 43, 1114–1122. [Google Scholar] [CrossRef] [PubMed]
  135. Yunus, U.; Tahir, M.K.; Moazzam Bhatti, M.H.; Ali, S.; Wong, W.-Y. 1-(1,3-Benzothiazol-2-yl)-3-benzoylthiourea. Acta Cryst. 2008, E64, o20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Saeed, S.; Rashid, N.; Hussain, R.; Jones, P.G. 1-(Benzothiazol-2-yl)-3-(4-nitrobenzoyl)- tiourea. Acta Cryst. 2009, E65, o2106. [Google Scholar]
  137. Siddiqui, N.; Rana, A.; Khan, S.A.; Syed Haque, S.E.; Alam, M.S.; Ahsan, W.; Ahmed, S. Synthesis of 8-substituted-4-(2/4-substituted phenyl)- 2H-[1,3,5]triazino[2,1-b][1,3]benzothiazole-2-thiones and their anticonvulsant, anti-nociceptive, and toxicity evaluation in mice. J. Enzyme Inhib. Med. Chem. 2009, 24, 1344–1350. [Google Scholar] [CrossRef]
  138. Modi, J.A.; Patel, K.C. Design, synthesis of some 2,6,9-trisubstituted purinyl thioureido derivatives and evaluation of antimicrobial activity. Med. Chem. Res. 2012, 21, 1660–1664. [Google Scholar] [CrossRef]
  139. Malik, S.; Bahare, R.D.; Khan, S.A. Design, synthesis and anticonvulsant evaluation of N-(benzo[d]thiazol-2-ylcarbamoyl)-2-methyl-4-oxoquinazoline-3(4H)-carbothioamide deri-vatives: A hybrid pharmacophore approach. Eur. J. Med. Chem. 2013, 67, 1–13. [Google Scholar] [CrossRef]
  140. Odame, F.; Woodcock, G.; Hosten, E.C.; Lobb, K.; Tshentu, Z.N. A novel gold(I)-mediated intramolecular transamidation of benzoyl thiourea derivatives to form benzamides via dethiocyanation. J. Organomet. Chem. 2020, 922, 121359. [Google Scholar] [CrossRef]
  141. Bhargava, P.N.; Lakhan, R. Synthesis of Benzothiazolylguanidines as Antituberculars and Antibacterials. Agric. Biol. Chem. 1968, 32, 1392–1394. [Google Scholar] [CrossRef]
  142. Bhargava, P.N.; Singh, H. New Compounds. Synthesis of Some New N-o-Tolyl-N’-2-(substituted) Benzothiazolylguanidines. J. Med. Chem. 1969, 12, 558–559. [Google Scholar] [CrossRef]
  143. Bhargava, P.N.; Choubey, V.N. Benzothiazolyl Guanidines as Antibacterials. Agric. Biol. Chem. 1970, 34, 644–647. [Google Scholar] [CrossRef]
  144. Bhargava, P.N.; Singh, S.N. Substituted Benzothiazolylguanidines. Curr. Sci. 1971, 40, 430–432. [Google Scholar]
  145. Lakhan, R.; Sharma, B.P.; Shukla, B.N. Synthesis and antimicrobial activity of 1-aryl-2-amino-3-(4-arylthiazol-2-yl)/(benzothiazol-2-yl)-guanidines. Il. Farm. 2000, 55, 331–337. [Google Scholar] [CrossRef] [PubMed]
  146. Venkatesh, P.; Tiwari, V.S. Design and synthesis of Quinazolinone, Benzothiazole derivatives bearing guanidinopropanoic acid moiety and their Schiff bases as cytotoxic and antimicrobial agents. Arabian J. Chem. 2016, 9, S914–S925. [Google Scholar] [CrossRef]
  147. Liu, Y.-H.; Cao, L.-H. Synthesis and bioactivity of novel methyl 6-deoxy-6-(N’-alkyl/aryl-N’’-benzothiazol-2-yl)-guanidino-α-D-glucopyranosides. Carbohydr. Res. 2008, 343, 615–625. [Google Scholar] [CrossRef] [PubMed]
  148. Anzini, M.; Chelini, A.; Mancini, A.; Cappelli, A.; Frosini, M.; Ricci, L.; Valoti, M.; Magistretti, J.; Castelli, L.; Giordani, A.; et al. Synthesis and biological evaluation of amidine, guanidine, and thiourea derivatives of 2-amino(6-trifluoromethoxy)-benzothiazole as neuroprotective agents potentially useful in brain diseases. J. Med. Chem. 2010, 53, 734–744. [Google Scholar] [CrossRef]
Figure 1. Biologically active urea- and thiourea-benzothiazole derivatives.
Figure 1. Biologically active urea- and thiourea-benzothiazole derivatives.
Ijms 24 09488 g001
Scheme 1. Synthesis of 6-substituted-BT-dihydrouracils 3a and 3b.
Scheme 1. Synthesis of 6-substituted-BT-dihydrouracils 3a and 3b.
Ijms 24 09488 sch001
Scheme 2. Synthesis of N-(4-methyl-BT-2-yl)-N-′(BI-N-sulfonyl)urea 4.
Scheme 2. Synthesis of N-(4-methyl-BT-2-yl)-N-′(BI-N-sulfonyl)urea 4.
Ijms 24 09488 sch002
Scheme 3. 1-(4/6-substituted-BT-2-yl)-3-phenyl-2-thiobarbituric acids 6 from condensation of N-(4/6-substituted-BT-2-yl)-N′-(phenyl)thioureas 5ai.
Scheme 3. 1-(4/6-substituted-BT-2-yl)-3-phenyl-2-thiobarbituric acids 6 from condensation of N-(4/6-substituted-BT-2-yl)-N′-(phenyl)thioureas 5ai.
Ijms 24 09488 sch003
Scheme 4. Transformation of nitrile to tetrazolyl group in N-(6-nitrile-BT-2-yl)-N′-(p-MeObenzyl)urea 8.
Scheme 4. Transformation of nitrile to tetrazolyl group in N-(6-nitrile-BT-2-yl)-N′-(p-MeObenzyl)urea 8.
Ijms 24 09488 sch004
Scheme 5. Substituted bis-BTs 10al from N-(4/6-substituted-BT-2-yl)-N′-(p-substituted-aryl)thioureas 9al.
Scheme 5. Substituted bis-BTs 10al from N-(4/6-substituted-BT-2-yl)-N′-(p-substituted-aryl)thioureas 9al.
Ijms 24 09488 sch005
Scheme 6. Ether cleavage of N-(6-methoxy-BT-2-yl)-N′-(3-metoxy-phenyl)-(thio)ureas 11a,b to afford the hydroxy-derivatives 12a,b.
Scheme 6. Ether cleavage of N-(6-methoxy-BT-2-yl)-N′-(3-metoxy-phenyl)-(thio)ureas 11a,b to afford the hydroxy-derivatives 12a,b.
Ijms 24 09488 sch006
Scheme 7. Substitution reactions of N-(5-benzyloxy-7-bromo-BT-2-yl)-N′-(ethyl)urea 13 to afford N-(5-pyrimidin-5-yl, 7-pyridin-2-yl-BT-2-yl)-N′-(ethyl)ureas 18ad, 19ah, and 20ad.
Scheme 7. Substitution reactions of N-(5-benzyloxy-7-bromo-BT-2-yl)-N′-(ethyl)urea 13 to afford N-(5-pyrimidin-5-yl, 7-pyridin-2-yl-BT-2-yl)-N′-(ethyl)ureas 18ad, 19ah, and 20ad.
Ijms 24 09488 sch007
Figure 2. Alcohol-containing N-(5-pyrimidin-5-yl-7-pyridin-2-yl-BT-2-yl)-N′-(ethyl)ureas 21as, 22ak, and 23af synthesized as Scheme 7.
Figure 2. Alcohol-containing N-(5-pyrimidin-5-yl-7-pyridin-2-yl-BT-2-yl)-N′-(ethyl)ureas 21as, 22ak, and 23af synthesized as Scheme 7.
Ijms 24 09488 g002aIjms 24 09488 g002b
Scheme 8. Synthesis of (Z)-ethyl-2-((Z)-2-(BT-2-ylimino)-4-oxo-3-phenylthiazolidin-5-ylidene)acetate 24 as receptor chemosensor.
Scheme 8. Synthesis of (Z)-ethyl-2-((Z)-2-(BT-2-ylimino)-4-oxo-3-phenylthiazolidin-5-ylidene)acetate 24 as receptor chemosensor.
Ijms 24 09488 sch008
Scheme 9. N-[6-(2,3-disubstituted-pyridin-5-yl)-BT-2-yl]-N′-(alkyl)ureas 27al from N-(6-bromo-BT-2-yl)-N′-(alkyl)ureas 25al.
Scheme 9. N-[6-(2,3-disubstituted-pyridin-5-yl)-BT-2-yl]-N′-(alkyl)ureas 27al from N-(6-bromo-BT-2-yl)-N′-(alkyl)ureas 25al.
Ijms 24 09488 sch009
Figure 3. Comparative N-[6-(5-substituted-6-methoxy-pyridin-3-yl)-BT-2-yl]-N′-[2-(morpholin-4-yl)-ethyl]urea 28mq.
Figure 3. Comparative N-[6-(5-substituted-6-methoxy-pyridin-3-yl)-BT-2-yl]-N′-[2-(morpholin-4-yl)-ethyl]urea 28mq.
Ijms 24 09488 g003
Scheme 10. Cyclization of N-(BT-2-yl)- N′-(Aryl)thioureas 29ag to 2-(N-thiazolidin-2-ene-4-one)-ABTs 30ag.
Scheme 10. Cyclization of N-(BT-2-yl)- N′-(Aryl)thioureas 29ag to 2-(N-thiazolidin-2-ene-4-one)-ABTs 30ag.
Ijms 24 09488 sch010
Scheme 11. Functionalization of N-(6-amino-BT-2-yl)-N′-(ethyl)urea 32 to afford N-(6-substituted-BT-2-yl)-N′-(ethyl)ureas 33a,b, 34ac, 35, 36a,b, and 37.
Scheme 11. Functionalization of N-(6-amino-BT-2-yl)-N′-(ethyl)urea 32 to afford N-(6-substituted-BT-2-yl)-N′-(ethyl)ureas 33a,b, 34ac, 35, 36a,b, and 37.
Ijms 24 09488 sch011
Scheme 12. Synthesis and cyclization reaction of N-(6-substituted-BT-2-yl)thioureas 39ad and 41ad.
Scheme 12. Synthesis and cyclization reaction of N-(6-substituted-BT-2-yl)thioureas 39ad and 41ad.
Ijms 24 09488 sch012
Scheme 13. Synthesis of 6-substituted-BT-thiosemicarbazones 4548.
Scheme 13. Synthesis of 6-substituted-BT-thiosemicarbazones 4548.
Ijms 24 09488 sch013
Scheme 14. Synthesis of 6-substituted-BT-semicarbazones 49ao.
Scheme 14. Synthesis of 6-substituted-BT-semicarbazones 49ao.
Ijms 24 09488 sch014
Scheme 15. Synthesis and cyclocondensation reaction of BT-thiosemicarbazones 51ac to 52a-c.
Scheme 15. Synthesis and cyclocondensation reaction of BT-thiosemicarbazones 51ac to 52a-c.
Ijms 24 09488 sch015
Scheme 16. 6-substituted-BT-(N-4-aryl-3-chloro-azetidinyl)semicarbazides 54at from N-(6-substituted-BT-2-yl)ureas 53at.
Scheme 16. 6-substituted-BT-(N-4-aryl-3-chloro-azetidinyl)semicarbazides 54at from N-(6-substituted-BT-2-yl)ureas 53at.
Ijms 24 09488 sch016
Scheme 17. 5-methyl-BT-thiosemicarbazones 55ac derived from N-(5-Methyl-BT-2-yl)thiourea and acetophenones.
Scheme 17. 5-methyl-BT-thiosemicarbazones 55ac derived from N-(5-Methyl-BT-2-yl)thiourea and acetophenones.
Ijms 24 09488 sch017
Scheme 18. Transformation of 6-substituted-BT-semicarbazides to N-(5-aminophenyl-[1,3,4]thiadiazol-2-yl)-2ABTs 56af.
Scheme 18. Transformation of 6-substituted-BT-semicarbazides to N-(5-aminophenyl-[1,3,4]thiadiazol-2-yl)-2ABTs 56af.
Ijms 24 09488 sch018
Scheme 19. Transformation of 6-chloro-BT-thiosemicarbazides 57 to N-(6-chloro-BT-2-yl)-N-(heterodiazol-2-yl)amines 5861.
Scheme 19. Transformation of 6-chloro-BT-thiosemicarbazides 57 to N-(6-chloro-BT-2-yl)-N-(heterodiazol-2-yl)amines 5861.
Ijms 24 09488 sch019
Scheme 20. 6-Chloro-BT-thiosemicarbazones 62ah derived from 6-chloro-BT-thiosemicarbazides and substituted benzaldehydes.
Scheme 20. 6-Chloro-BT-thiosemicarbazones 62ah derived from 6-chloro-BT-thiosemicarbazides and substituted benzaldehydes.
Ijms 24 09488 sch020
Figure 4. 6-Nitro-BT-semicarbazones 63au and 64ad as the rat MAO-B isoenzime inhibitors.
Figure 4. 6-Nitro-BT-semicarbazones 63au and 64ad as the rat MAO-B isoenzime inhibitors.
Ijms 24 09488 g004
Scheme 21. N-(6-fluoro,7-chloro-BT-2-yl)-N′-(2-aryl-4-one-thiazolidin-3-yl)ureas 68ae derived from the corresponding disubstituted-BT-semicarbazide 66 and substituted benzaldehydes.
Scheme 21. N-(6-fluoro,7-chloro-BT-2-yl)-N′-(2-aryl-4-one-thiazolidin-3-yl)ureas 68ae derived from the corresponding disubstituted-BT-semicarbazide 66 and substituted benzaldehydes.
Ijms 24 09488 sch021
Scheme 22. 6-substituted-BT-semicarbazones 70ah, 71ax, and 72ad from the reaction of 6-substituted-BT-semicarbazides 69ae with o-hydroxybenzaldehydes.
Scheme 22. 6-substituted-BT-semicarbazones 70ah, 71ax, and 72ad from the reaction of 6-substituted-BT-semicarbazides 69ae with o-hydroxybenzaldehydes.
Ijms 24 09488 sch022
Figure 5. 6-substituted-BT-semicarbazones with ortho-hydroxy-N-acylhydrazone moiety 73af, 74aq and 75ac as potent antitumor agents.
Figure 5. 6-substituted-BT-semicarbazones with ortho-hydroxy-N-acylhydrazone moiety 73af, 74aq and 75ac as potent antitumor agents.
Ijms 24 09488 g005
Figure 6. 6-substituted-BT-semicarbazones with indole-based moiety 76ac, 77ae, and 78azi as potent antitumor agents.
Figure 6. 6-substituted-BT-semicarbazones with indole-based moiety 76ac, 77ae, and 78azi as potent antitumor agents.
Ijms 24 09488 g006
Scheme 23. N-(6-cloro-BT-2-yl) N′-(thiazolidinyl-4-one)ureas 80ag and N-(6-chloro-BT-2-yl)-N′-(4-aryl-3-chloro-azetidinyl-2-one)ureas 81ag from 6-chloro-BT-semicarbazones 79ag.
Scheme 23. N-(6-cloro-BT-2-yl) N′-(thiazolidinyl-4-one)ureas 80ag and N-(6-chloro-BT-2-yl)-N′-(4-aryl-3-chloro-azetidinyl-2-one)ureas 81ag from 6-chloro-BT-semicarbazones 79ag.
Ijms 24 09488 sch023
Scheme 24. 6-Substituted-BT-semicarbazones 82al derived from 6-substituted-BT-semicarbazides and heteroaldehydes or heteroketones.
Scheme 24. 6-Substituted-BT-semicarbazones 82al derived from 6-substituted-BT-semicarbazides and heteroaldehydes or heteroketones.
Ijms 24 09488 sch024
Scheme 25. 6/4-Substituted-BT-semicarbazone-bis-sulfoxides 84al from oxidation of the corresponding bi-sulfides 83al derived from the 6/4-substituted-BT-semicarbazides.
Scheme 25. 6/4-Substituted-BT-semicarbazone-bis-sulfoxides 84al from oxidation of the corresponding bi-sulfides 83al derived from the 6/4-substituted-BT-semicarbazides.
Ijms 24 09488 sch025
Scheme 26. N-(BT-2-yl)-N′-(substituted)(thio)ureas 8689 from 1,1′-(thio)carbonyl-BT-imidazoles 85a,b.
Scheme 26. N-(BT-2-yl)-N′-(substituted)(thio)ureas 8689 from 1,1′-(thio)carbonyl-BT-imidazoles 85a,b.
Ijms 24 09488 sch026
Scheme 27. N-(6/5-substituted-BT-2-yl)-N′-(benzoyl)thioureas 90ak from 6/5-substituted-2ABTs and benzoylthiocyanates.
Scheme 27. N-(6/5-substituted-BT-2-yl)-N′-(benzoyl)thioureas 90ak from 6/5-substituted-2ABTs and benzoylthiocyanates.
Ijms 24 09488 sch027
Figure 7. N-(6-Substituted-BT-2-yl)-N′-(benzoyl)thioureas 91at from 6-substituted-2ABTs and substituted benzoylthiocyanates.
Figure 7. N-(6-Substituted-BT-2-yl)-N′-(benzoyl)thioureas 91at from 6-substituted-2ABTs and substituted benzoylthiocyanates.
Ijms 24 09488 g007
Figure 8. Intramolecular hydrogen bonding in N-(BT-2-yl)- N′-(benzoyl)thioureas 90a and 92.
Figure 8. Intramolecular hydrogen bonding in N-(BT-2-yl)- N′-(benzoyl)thioureas 90a and 92.
Ijms 24 09488 g008
Scheme 28. Cyclization reaction of N-(6-substituted-BT-2-yl)—N′-(benzoyl)thioureas 93at.
Scheme 28. Cyclization reaction of N-(6-substituted-BT-2-yl)—N′-(benzoyl)thioureas 93at.
Ijms 24 09488 sch028
Scheme 29. N-(substituted-BT-2-yl)-N′-(purinoacetyl)thioureas 95aj from substituted-2ABT and purinoacetyl-isothiocyanate.
Scheme 29. N-(substituted-BT-2-yl)-N′-(purinoacetyl)thioureas 95aj from substituted-2ABT and purinoacetyl-isothiocyanate.
Ijms 24 09488 sch029
Scheme 30. N-(5-substituted-BT-2-yl)-N′-(acyloyl)thioureas 96aade derived from 5-substituted-2ABTs and acyloyl-isothiocyanates.
Scheme 30. N-(5-substituted-BT-2-yl)-N′-(acyloyl)thioureas 96aade derived from 5-substituted-2ABTs and acyloyl-isothiocyanates.
Ijms 24 09488 sch030
Scheme 31. N-(substituted-BT-2-yl)-N′-[(2-methyl-4-oxoquinazolinethioyl-3(4H)]thioureas 98–100aj derived from N-(substituted-BT-2-yl)-N′-(thiocarbamidoyl)thioureas 97aj.
Scheme 31. N-(substituted-BT-2-yl)-N′-[(2-methyl-4-oxoquinazolinethioyl-3(4H)]thioureas 98–100aj derived from N-(substituted-BT-2-yl)-N′-(thiocarbamidoyl)thioureas 97aj.
Ijms 24 09488 sch031
Scheme 32. Conversion of N-(BT-2-yl)-N′-(benzoyl)thioureas 101ai to 2-(substituted-benzamide)-BTs 102ai.
Scheme 32. Conversion of N-(BT-2-yl)-N′-(benzoyl)thioureas 101ai to 2-(substituted-benzamide)-BTs 102ai.
Ijms 24 09488 sch032
Scheme 33. Amination-desulfurization of N-(substituted-BT-2-yl)-N′-(p-tolyl)thioureas 103 to afford N-(substituted-BT-2-yl)-N′-(p-tolyl)-N″-(alkyl)guanidines 104106.
Scheme 33. Amination-desulfurization of N-(substituted-BT-2-yl)-N′-(p-tolyl)thioureas 103 to afford N-(substituted-BT-2-yl)-N′-(p-tolyl)-N″-(alkyl)guanidines 104106.
Ijms 24 09488 sch033
Figure 9. N-(substituted-BT-2-yl)-N′-(p-chlorophenyl)-N″-(alkyl)guanidines 107111.
Figure 9. N-(substituted-BT-2-yl)-N′-(p-chlorophenyl)-N″-(alkyl)guanidines 107111.
Ijms 24 09488 g009
Figure 10. N-(substituted-BT-2-yl)-N′-(benzyl)guanidines 112ak.
Figure 10. N-(substituted-BT-2-yl)-N′-(benzyl)guanidines 112ak.
Ijms 24 09488 g010
Figure 11. N-(substituted-BT-2-yl)-N′-(p-bromophenyl)-N″-(alkyl)guanidines 113a-k and 114a-k.
Figure 11. N-(substituted-BT-2-yl)-N′-(p-bromophenyl)-N″-(alkyl)guanidines 113a-k and 114a-k.
Ijms 24 09488 g011
Scheme 34. Amination-desulfurization of N-(BT-2-yl)-N′-(aryl)thioureas to afford the corresponding GBTs 115ad.
Scheme 34. Amination-desulfurization of N-(BT-2-yl)-N′-(aryl)thioureas to afford the corresponding GBTs 115ad.
Ijms 24 09488 sch034
Scheme 35. N-(6-substituted-BT-2-yl)-N′-(2-carboxy-ethanoyl)guanidines 117af, from amination-desulfurization of N-(6-substituted-BT-2-yl)thioureas 116af.
Scheme 35. N-(6-substituted-BT-2-yl)-N′-(2-carboxy-ethanoyl)guanidines 117af, from amination-desulfurization of N-(6-substituted-BT-2-yl)thioureas 116af.
Ijms 24 09488 sch035
Scheme 36. N-(6-substituted-BT-2-yl)-N′(sugar)-N″-(alkyl)guanidines 119124 from amination-desulfurization of the N-(6-substituted-BT-2-yl)-N′-(sugar)thioureas 118ac.
Scheme 36. N-(6-substituted-BT-2-yl)-N′(sugar)-N″-(alkyl)guanidines 119124 from amination-desulfurization of the N-(6-substituted-BT-2-yl)-N′-(sugar)thioureas 118ac.
Ijms 24 09488 sch036
Scheme 37. Amination-desulfurization of N-(6-trifluoromethoxy-BT-2-yl)-N′-(alkyl)thioureas 125a,b through the in situ intermediate S-methylisothiourea-BT.
Scheme 37. Amination-desulfurization of N-(6-trifluoromethoxy-BT-2-yl)-N′-(alkyl)thioureas 125a,b through the in situ intermediate S-methylisothiourea-BT.
Ijms 24 09488 sch037
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mendieta-Wejebe, J.E.; Rosales-Hernández, M.C.; Padilla-Martínez, I.I.; García-Báez, E.V.; Cruz, A. Design, Synthesis and Biological Activities of (Thio)Urea Benzothiazole Derivatives. Int. J. Mol. Sci. 2023, 24, 9488. https://doi.org/10.3390/ijms24119488

AMA Style

Mendieta-Wejebe JE, Rosales-Hernández MC, Padilla-Martínez II, García-Báez EV, Cruz A. Design, Synthesis and Biological Activities of (Thio)Urea Benzothiazole Derivatives. International Journal of Molecular Sciences. 2023; 24(11):9488. https://doi.org/10.3390/ijms24119488

Chicago/Turabian Style

Mendieta-Wejebe, Jessica E., Martha C. Rosales-Hernández, Itzia I. Padilla-Martínez, Efrén V. García-Báez, and Alejandro Cruz. 2023. "Design, Synthesis and Biological Activities of (Thio)Urea Benzothiazole Derivatives" International Journal of Molecular Sciences 24, no. 11: 9488. https://doi.org/10.3390/ijms24119488

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop